EP4081537A1 - Neue mesothelin-spezifische chimäre antigen-rezeptoren (car) für die krebsimmuntherapie bei soliden tumoren - Google Patents

Neue mesothelin-spezifische chimäre antigen-rezeptoren (car) für die krebsimmuntherapie bei soliden tumoren

Info

Publication number
EP4081537A1
EP4081537A1 EP20845393.6A EP20845393A EP4081537A1 EP 4081537 A1 EP4081537 A1 EP 4081537A1 EP 20845393 A EP20845393 A EP 20845393A EP 4081537 A1 EP4081537 A1 EP 4081537A1
Authority
EP
European Patent Office
Prior art keywords
cells
car
mesothelin
cell
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP20845393.6A
Other languages
English (en)
French (fr)
Inventor
Cécile Schiffer-Mannioui
Philippe Duchateau
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cellectis SA
Original Assignee
Cellectis SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cellectis SA filed Critical Cellectis SA
Publication of EP4081537A1 publication Critical patent/EP4081537A1/de
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/54Pancreas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • CAR chimeric antigen receptors
  • the present invention relates to the field of cell immunotherapy and more particularly to engineered immune cells expressing new mesothelin (MLSN) specific chimeric antigen receptors (anti-mesothelin CAR) useful in the treatment of solid tumors.
  • MLSN new mesothelin
  • anti-mesothelin CAR anti-mesothelin CAR
  • Chimeric antigen receptors are synthetic receptors that target T cells to cell- surface antigens and augment T-cell function and persistence.
  • Mesothelin is a cell-surface antigen implicated in tumor invasion, which is highly expressed in mesothelioma and lung, pancreas, breast, ovarian, and other cancers.
  • Encouragingly, recent clinical trials evaluating active immunization or immunoconjugates in patients with pancreatic adenocarcinoma or mesothelioma have shown responses without toxicity.
  • these findings and preclinical CAR therapy models using either systemic or regional T-cell delivery argue favorably for mesothelin CAR therapy in multiple solid tumors.
  • Solid tumor CAR targets under investigation are altered gene products mostly arising from genetic mutations or altered splicing (EGFRvlll), altered glycosylation patterns (MUC1), cancer-testis antigen-derived peptides (MAGE), overexpressed differentiation antigens CEA, PSMA, GD2, MUC16, HER2/ERBB2, and mesothelin (MSLN), or tumor-associated stroma (FAP and VEGFR).
  • EGFRvlll genetic mutations or altered splicing
  • MUC1 glycosylation patterns
  • MAGE cancer-testis antigen-derived peptides
  • CEA cancer-testis antigen-derived peptides
  • PSMA cancer-testis antigen-derived peptides
  • MUC16 cancer-testis antigen-derived peptides
  • MSLN mesothelin
  • FAP and VEGFR tumor-associated stroma
  • an optimal solid-tumor antigen target is one whose expression either is restricted to tumor cells or occurs only at very low levels in expendable normal tissues.
  • MSLN has emerged as an attractive target for cancer immunotherapy, considering its low expression on normal mesothelial cells and high expression in a broad spectrum of solid tumors.
  • MSLN is a potential CAR target in a number of common solid tumors, such as at least oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, Lung cancer, Thymic carcinoma, mesothelioma, ovarian cancer, and endometrial cancer [Morello, A. et al. (2016) Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov. 6(2); 133-46]
  • MSLN is a glycoprotein anchored to the plasma membrane by a glycophosphatidyl inositol (GPI) domain. It is initially synthesized as a 69 kDa cell-surface protein. After cleavage of the amino terminus by the furin protease, a 40-kDa C-terminal fragment remains attached to the membrane and a soluble 32-kDa N-terminal fragment, named megakaryotic-potentiating factor (MPF), is released [Pastan, I., Hassan, R. (2014) Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer. Res.
  • GPI glycophosphatidyl inositol
  • SMRP soluble MSLN-related protein
  • MSLN The biologic function of MSLN seems to be nonessential in normal tissues, given that MSLN knockout mice exhibit normal development, reproduction, and blood cell count. In contrast, preclinical and clinical studies increasingly show that aberrant MSLN expression plays an active role in both malignant transformation of tumors and tumor aggressiveness by promoting cancer cell proliferation, contributing to local invasion and metastasis, and conferring resistance to apoptosis induced by cytotoxic agents.
  • MSLN can act bidirectionally, either by directly activating intracellular pathways via its GPI domain or by interacting with its receptor, CA125/MUC16. Overexpression of MSLN alone is sufficient to constitutively activate the NFKB, MAPK, and PI3K intracellular pathways promoting cell proliferation and resistance to apoptosis.
  • MSLN is expressed on mesothelial cells of the peritoneal and pleural cavities and pericardium; it is expressed minimally on the epithelial cell surface of the trachea, ovaries, rete testis, tonsils, and fallopian tubes.
  • Overexpression of MSLN was initially observed in mesothelioma and ovarian cancer, and subsequently in lung, esophageal, pancreatic, gastric, biliary, endometrial, thymic, colon, and breast cancers. MSLN overexpression thus has an estimated incidence of 340,000 patients and prevalence of 2 million patients a year in the United States alone.
  • CARs consist of an ectodomain commonly derived from a single-chain variable fragment (scFv), a hinge, a transmembrane domain, and an endodomain (typically comprising signaling domains derived from and costimulatory receptors). Second-generation CARs further enhance T-cell function and persistence through the incorporation of signalling domains that rescue and amplify the activation signal provided by the cytoplasmic domain. Dual signalling prevents T-cell anergy and increases persistence and function by augmenting T-cell proliferation and cytokine production (IFNy and IL2) and reducing activation-induced cell death through the recruitment of the PI3K, TRAF, and/or other pathways.
  • scFv single-chain variable fragment
  • a hinge typically comprising signaling domains derived from and costimulatory receptors
  • endodomain typically comprising signaling domains derived from and costimulatory receptors.
  • Second-generation CARs further enhance T-cell function and persistence through the incorporation of signal
  • Third-generation CARs comprise three signalling domains, typically encompassing those of and two co stimulatory domains, for example CD28 and 4-1 BB or CD28 and 0X40. Compared with second-generation CARs, third-generation CARs have shown inconsistent antitumor activity in vivo. Choosing an appropriate co-stimulation domain is essential to sustain CAR T-cell activity and calibrate T-cell persistence. However, the ideal co-stimulation domain may depend on context, as CAR function depends on multiple extraneous factors, such as antigen density, CAR stoichiometry, CAR affinity and the immunologic features of the tumor microenvironment.
  • the spatial distance between CARs and their target antigens may be equally important for effective initiation of T cell signalling, but depends on an entirely different set of structural elements related to the location of the epitope on the target molecule and the spacer domain between the scFv and the T cell membrane.
  • Several studies have demonstrated that the same epitope can activate CAR-T cells with greater efficiency when expressed in a more membrane- proximal position than a membrane-distal position. For example, Hombach et al.
  • CAR T cells recognizing the membrane-distal “N” epitope of the carcinoembryonic antigen (CEA) were only modestly activated; however, when they engineered recombinant CEA protein to express the N epitope in a membrane-proximal position, the same CAR T cells were activated more efficiently [Hombach AA, et a/.( 2007) T cell activation by antibody-like immunoreceptors: the position of the binding epitope within the target molecule determines the efficiency of activation of redirected T cells. J Immunol.
  • MSLN CAR T-cell activation (cytokine secretion and cytotoxic activity) seems to remain dependent on MSLN expression on the cell surface [Carpenito C., et al. (2009) Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. PNAS. 106:3360-5]
  • solid-tumor microenvironment poses several obstacles for MSLN CAR-T cells that may limit their antitumor efficacy.
  • CAR T cells To optimize the efficiency of CAR T cells, numerous approaches are under evaluation to tame the host tumor microenvironment or generate “armored” CAR T cells that can overcome immune barriers.
  • Such strategies include (i) promoting CAR T-cell infiltration, (ii) augmenting the functional persistence of CAR T cells, (iii) enhancing CAR T cells to overcome inhibitory signals encountered in the tumor microenvironment, and (iv) improving safety by preventing on-target/off-tumor toxicity.
  • combining specific CAR structures with gene edited cells appear to be most promising. Riese et al.
  • CAR T cells can be eliminated by drug-induced activation of a suicide gene, such as the herpes simplex thymidine kinase ( HSV-TK ), gene inducible caspase-9, or gene.
  • HSV-TK herpes simplex thymidine kinase
  • caspase-9 gene inducible caspase-9
  • the present invention aims to address all or part of the above limitations by providing safer engineered immune CAR positive cells to target MSLN expressing cells in-vivo, such as solid tumors, with the perspective of being used in allogeneic treatment strategies. Summary of the invention
  • the present invention primarily concerns mesothelin specific chimeric antigen receptors (CAR) for their expression into immune cells, preferably T-cells, for their therapeutic use against malignant mesothelin expressing cells or tissues.
  • CAR mesothelin specific chimeric antigen receptors
  • Such CARs present a structure comprising typically:
  • cytoplasmic domain comprising a CD3 zeta signalling domain and a co-stimulatory domain.
  • the extracellular ligand binding domain of the CARs according to the present invention preferably comprises one or several scFv segment from the antibody referred to as mesol , and more particularly the CDRs therefrom including SEQ ID NO:3, 4, 5, 6, 7 and/or 8.
  • said extracellular ligand binding domain of said CARs comprise:
  • variable heavy VH chain comprising CDRs from the antibody mesol having respectively at least 90% identity with SEQ ID NO:3 (CDRH1-Meso1), SEQ ID NO:4 (CDRH2- mesol) and/or SEQ ID NO:5 (CDRH3-meso1), and
  • variable heavy VL chain comprising CDRs from the antibody Mesol having respectively at least 90% identity with SEQ ID NO:6 (CDRL1- mesol), SEQ ID NO:7 (CDRL2- mesol) and/or SEQ ID NO:8 (CDRL3- mesol).
  • the anti-mesothelin CARs of the invention form exogenous polypeptide sequences, which are expressed by the immune cells for their exposition at the surface of the cells.
  • polynucleotide sequences exogenous preferably inserted at specific genomic loci, such as at the TCR, B2m or PD1 gene loci by using rare-cutting endonucleases.
  • said CARs further comprise additional exogenous polypeptide sequence including epitopes, which can be targeted by clinically approved antibodies for in-vivo depletion or by other ligands for their in-vivo or in-vitro detection or purification.
  • additional exogenous polypeptide segment may be specifically recognized by Rituximab, such as that referred to as “R2” in the present specification.
  • the invention is more particularly drawn to immune cells or populations of immune cells transformed with the anti-mesothelin CARs polynucleotide sequences comprising said polynucleotide sequences and/or expressing polypeptide anti-mesothelin CARs sequences.
  • Such engineered immune cells, or populations of cells, according to the invention may be further genetically engineered, mutated or gene-edited to improve their therapeutic suitability or potency, such as to improve their persistence or lifespan.
  • the engineered immune cells according to the invention combine the expression of anti-mesothelin CARs sequences with other genetic modifications reducing the expression of their endogenous genes, such as TCR, HLA, and/or B2m genes.
  • TGFbeta receptor such as TCR, HLA, and/or B2m genes.
  • the engineered immune cells can be mutated to improve their CAR-dependent immune activation, in particular by reducing or suppressing the expression of immune checkpoint proteins and/or receptors thereof, such as PD1/ PDL1.
  • the engineered immune cells can be mutated to improve their CAR-dependent immune activation, in particular by reducing or suppressing TGFbeta signalling pathway.
  • further exogenous genetic sequences can also be inserted, co-transfected or co-expressed with the anti-mesothelin CARs of the present invention, in particular inhibitors or decoy of TGFbeta receptor, such as a dominant negative TGFbeta receptor (dnTGFbRII).
  • TGFbeta receptor such as a dominant negative TGFbeta receptor (dnTGFbRII).
  • exogenous genetic sequences which expression can be combined with that of the anti-mesothelin CARs to improve therapeutic potency of immune cells, in particular the following ones:
  • - NK cell inhibitors such as HLAG, HLAE or ULBP1;
  • - CRS inhibitors such as a mutated IL6Ra, sGP130 or IL18-BP; or
  • DHFR Dihydrofolate reductase
  • IMPDH2 inosine monophosphate dehydrogenase 2
  • MGMT calcineurin or methylguanine transferase
  • mTORmut conferring drug resistance
  • cytokine such as IL-2, IL-12 and IL-15.
  • TAM Tumor Associated Macrophages
  • the engineered immune cells according to the present invention are particularly suited for treating a condition characterized by mesothelin expressing cells, in particular solid tumors, such as typically: oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, lung cancer, thymic carcinoma, mesothelioma, ovarian cancer and/or endometrial cancer.
  • solid tumors such as typically: oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, lung cancer, thymic carcinoma, mesothelioma, ovarian cancer and/or endometrial cancer.
  • the invention thus encompasses methods for producing engineered cells, the resulting therapeutic cells, populations of cells comprising such cells and therapeutic compositions comprising same, as well as the methods of treatment allowing to address pathologies induced by mesothelin expressing cells.
  • FIG. 1 Structural representation of preferred versions of anti-mesothelin CAR according to the invention.
  • A CAR comprising: V1 and V2 representing sequences comprising ScFv specifically binding mesothelin, such as VH and VL or VL and VH from mesol antibody; L: linker; R1 and R2 representing exogenous epitopes such as CD20 epitopes recognized by human approved monoclonal anti CD20 antibodies (e.g.: rituximab,...); TM: transmembrane domain; CO-STIM: co-stimulatory domain; ITAMs: stimulatory domain comprising ITAM (Immunoreceptor tyrosine-based activation motif (ITAM).
  • ITAM Interleukin activation motif
  • Such CAR has typically at least 80% polypeptide sequence identity with SEQ ID NO:21.
  • B CAR without exogenous epitopes comprising: VH and VL or VL and VH from mesol antibody; a (G4S)3 linker; a CD8a hinge domain a CD8a transmembrane domain; a 4-1 BB co-stimulatory domain; and a CD3z signaling domain.
  • FIG. 2 Schematic representation figuring anti-MSLN CAR expressing immune cells according to the invention, in which optional genetic attributes have been further introduced :
  • A Co-expression with an inactive variant of TGF ⁇ receptor (e.g.: dnTGFbRII) and/or genetic reduction or inactivation of TGF ⁇ receptor expression to counteract tumor-induced immune suppression. Reduction or inactivation of TCR expression (e.g. TCRalpha) to lower immune T-cells alloreactivity causing GvHD.
  • B Genetic reduction or inactivation via gene editing tools (such as TALEN) of TGF ⁇ receptor, TCR and/or CD52 expression.
  • FIG 3 Mesothelin protein expression at the surface of 293H, A2058, HeLa, and HPAC cells. Analysis of MSLN expression was performed by flow cytometry using a mouse monoclonal anti-human MSLN antibody as primary antibody and the APC-conjugated goat anti-mouse polyclonal antibody as secondary antibody.
  • Figure 4 Quantitative Mesothelin protein expression at the surface of HeLa, and HPAC cells. Analysis of MSLN expression level was performed by flow cytometry using fluorescence-based QIFIKIT.
  • Figure 5 Figure representing serial killing assay performed to assess in-vitro activation of anti- mesothelin CAR positive cells.
  • FIG. 6 CAR expression at the surface of primary [TCRalpha] neg T-cells (UCART cells).
  • Cryopreserved UCART cells generated from a single donor were stained with histidine-tagged recombinant human mesothelin protein and PE-conjugated anti-histidine antibody or biotinylated protein L and Vioblue-conjugated streptavidin, and analyzed by flow cytometry.
  • Figure 7 CD4 and CD8 expression by the CAR+ fraction of UCART cells.
  • Cryopreserved UCART cells generated from a single donor were stained with FITC-conjugated anti-CD4 and BV510-conjugated anti CD8 antibodies and analyzed by flow cytometry.
  • FIG. 8 Production of IFNg by UCART cells. Fresh UCART cells generated from a single donor were cocultured for 24 hours with (A) HPAC (MSLN+) cells, (B) A2058 (MSLN-) cells, and (C) 293H (MSLN-) cells. IFNg produced in the culture supernatants was quantified by ELISA.
  • FIG. 9 Graphs showing % cell lysis resulting from the serial killing assay of HPAC cells by primary [TCRalpha] neg T-cells (UCART cells), which protocol is illustrated in Figure 5. Cryopreserved UCART cells generated from one donor were cocultured for 15 days with HPAC cells at E:T ratios of 1 :2 (A) or 1:8 (B).
  • FIG. 10 TCRap expression at the UCART cells surface. Cryopreserved UCART cells generated from a single donor were stained with PEvio770-conjugated anti-TCRap antibody and analyzed by flow cytometry.
  • FIG. 11 Flow cytometry analysis of the expression of the TCRap receptor at the surface of non-engineered T cells, T cells knocked-out for the TRAC gene, and T cells knocked-out for the TRAC gene and depleted in TCRap + cells.
  • Figure 12 Flow cytometry analysis of the expression of CD25 at the surface of non-engineered T-cells, T-cells knocked-out for the TRAC gene and T-cells knocked-out for the TRAC gene and depleted in TCRap+ cells, upon exposure to medium (red line), + 0.1 ⁇ g/ml of PHA-L (orange line), + 0.25 ⁇ g/ml of PHA-L (green line) and + 2.5 ⁇ g/ml of PHA-E and L (blue line) respectively.
  • Figure 13 Measurement of UCART cells depletion by rituximab-mediated CDC through the exogenous epitope polypeptide R2 included into the CARs P4-R2, Meso1-R2 and MES02-R2.
  • Figure 14 Graphs displaying measurement of SMAD2-3 phosphorylation upon exposure to TGF ⁇ of CART cells generated from 2 different donors.
  • Figure 15 Mean tumor volume (HPAC MSLN+ cells) in mice injected with UCARTmeso (TCR negative anti-mesolthelin P4-R2 CAR positive cells) at three doses (1x10 6 , 3x10 6 and 10x10 6 CAR+ cells/mouse).
  • Figure 16 Mean tumor volume in mice injected with UCARTmeso cells (TCR negative anti- mesolthelin Meso2-R2 CAR positive cells) at three doses (1x10 6 , 3x10 6 and 10x10 6 CAR+ cells/mouse).
  • Figure 17 Mean tumor volume in mice injected with UCARTmeso cells (TCR negative anti- mesolthelin Meso1-R2 CAR positive cells) at three doses (1x10 6 , 3x10 6 and 10x10 6 CAR+ cells/mouse).
  • Figure 18 Mean tumor volume +/- standard deviation in mice injected with UCARTmeso cells at three doses (A:1x10 6 , B:3x10 6 and C:10x10 6 CAR+ cells/mouse). Comparison of the different doses for respectively Meso1-R2, P4-R2 and MES02-R2.
  • Figure 19 Mean tumor volume in mice injected with UCARTmeso cells that are also expressing dnTGFBRII at two doses (3x10 6 and 10x10 6 CAR+ cells/mouse).
  • Figure 20 A. Comparison of CAR and dnTGFBRII detection in UCARTMeso cells expressing P4 or MES01 constructs with dnTGFBRII. B. Percentage of CD4+ and CD8+ in the CAR positive fraction of UCARTMeso cells produced in example 5.
  • Figure 21 Percentage of Temra, Tem, Tcm; Tn/scm cells observed in the CAR+ CD4+ fraction (A.) or CAR+ CD8+ fraction (B.) of UCARTMeso cells produced in example 5.
  • Figure 22 Percentage of H226 cells killing by different UCARTmeso cells inactivated or not for TGFBRII pathway by Knock-Out (KO) or by expression of dominant negative TGFBRII (dnTGFBRII).
  • Figure 23 Production of IFNg by UCARTMeso cells produced in example 5 and exposed (A.) or not (B.) to recombinant Mesothelin protein.
  • Figure 24 Evaluation of UCARTmeso cells sensitivity to TGFb.
  • A Percentage of pSMAD2/3 positive (grey) or negative (black) cells among the CAR positive fraction of UCARTmeso produced in example5 upon treatment with TGFb.
  • B Percentage of proliferation inhibition of the different UCARTmeso in presence of TGFb and recombinant mesothelin protein.
  • Table 1 Amino acid sequence of the different domains constituting the P4, Mesd and MES02 scFvs of the CAR illustrated in the examples.
  • Table 2 Amino acid sequence of the different domains, others than the scFv, constituting the MSLN CARs according to the invention.
  • Table 3 Examples of mAb-specific epitopes (and their corresponding mAbs) which can be used in the extracellular binding domain of the CAR of the invention for engineered cells sorting and depletion.
  • Table 4 Amino acid sequences of P4-R2, Meso1-R2, Mesd and MES02-R2 CARs.
  • Table 5 Examples of mAb-specific epitopes (and their corresponding mAbs), which can be inserted in the extracellular binding domain of the CAR of the invention.
  • Table 6 TALE-nuclease target sequences for TGF ⁇ RII gene.
  • Table 8 Genomic sequences targeted by the TALE-nucleases (TALEN) to inactivate TCR and CD52.
  • Table 9 Characteristics of the population of engineered T-cells used in the examples.
  • Table 10 Description of the six types of genetically modified T cells generated for the study provided in example 5.
  • the present invention is drawn to a general method of treating solid tumors by adoptive immune cells directed against the transmembrane protein MSLN, in particular the mesothelin’s specific epitope region spanning the polypeptide sequence SEQ ID NO:25 in this protein, and more particularly by using allogeneic CAR-T cells directed against this epitope, which have proven particular efficiency.
  • NK or T- cells armed with a CAR comprising SEQ ID NO:9 and/or SEQ ID NO: 10 have shown higher activation, potency, killing activity, cytokine release, and in-vivo persistence than their counterparts endowed with other prior anti-mesothelin CARs.
  • the present invention thus is drawn to CAR immune cells targeting specific epitope(s) comprised in the sequence SEQ ID NO.25 of MSLN protein, which is present at the surface of malignant cells, especially engineered for treating solid tumors.
  • CAR Chimeric Antigen Receptor
  • scFv single-chain antibody
  • Binding moieties based on receptor or ligand domains have also been used successfully.
  • the signaling domains of CARs are generally derived from the cytoplasmic region of the CD3zeta or the Fc receptor gamma chains, which are generally combined with signaling domains from co-stimulatory molecules including CD28, OX-40 (CD134), ICOS and 4-1 BB (CD137) to enhance survival and increase proliferation of the cells.
  • CARs are generally expressed in effector immune cells to redirect their immune activity against antigens expressed at the surface of tumor cells from various malignancies including lymphomas and solid tumors.
  • a component of a CAR is any functional subunit of a CAR that is encoded by an exogenous polynucleotide sequence introduced into the cell. For instance, this component can help to interact with the target antigen, the stability or the localization of the CAR into the cell.
  • such CAR comprises:
  • a signal transducing domain preferably a cytoplasmic domain comprising a CD3 zeta signalling domain and a co-stimulatory domain.
  • the present invention is drawn more particularly to CARs, which are expressed in immune cells, such as NK or T-cells, said CAR comprising an antigen binding domain specifically binding SEQ ID NO:25.
  • the mesothelin specific chimeric antigen receptor (CAR) of the present invention has an extra cellular ligand binding-domain comprising at least one CDR region from the variable heavy VH chain of the antibody Mesol , selected from CDRH1-Meso1 (having identity with SEQ ID NO:3), CDRH2-Meso1 (having identity with SEQ ID NO:4) and CDRH3-Meso1 (having identity with SEQ ID NO:5, and/or from the variable heavy VL chain of said antibody selected from CDRL1-Meso1 (having identity with SEQ ID NO:6), CDRL2-Meso1 (having identity with SEQ ID N0:7), and CDRL3-Meso1 (having identity with SEQ ID N0:8).
  • CDRH1-Meso1 having identity with SEQ ID NO:3
  • CDRH3-Meso1 having identity with SEQ ID NO:5
  • said extra cellular ligand binding-domain comprises:
  • variable heavy VH chain comprising CDRs from the antibody Mesol having respectively at least 90% identity with SEQ ID NO:3 (CDRH1-Meso1), SEQ ID NO: 4 (CDRH2- Mesol) and SEQ ID NO:5 (CDRH3-Meso1), and/or
  • variable heavy VL chain comprising CDRs from the antibody Mesol having respectively at least 90% identity with SEQ ID NO:6 (CDRL1-Meso1), SEQ ID NO:7 (CDRL2- Mesol) and SEQ ID NO:8 (CDRL3-Meso1).
  • the mesothelin specific chimeric antigen receptor has an extra cellular ligand binding-domain, which comprises VH and VL chains having at least 80%, preferably at least 90%, more preferably at least 95%, and even more preferably at least 99% sequence identity respectively with SEQ ID NO:9 (Mesol -VH) and SEQ ID NO:10 (Meso1-VL).
  • VH and VL chains having at least 80%, preferably at least 90%, more preferably at least 95%, and even more preferably at least 99% sequence identity respectively with SEQ ID NO:9 (Mesol -VH) and SEQ ID NO:10 (Meso1-VL).
  • framework residues in the framework regions can be substituted with the corresponding residue from the CDR donor antibody to alter, for example improve, antigen binding.
  • framework substitutions are identified by methods well-known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions [See, e.g., Queen et al., U.S. Pat. No. 5,585,089; and Riechmann et al., (1988) Nature , 332:323, which are incorporated herein by reference in their entireties].
  • Table 1 Amino acid sequence of the different domains constituting the P4, Meso1 and MES02 scFvs.
  • Table 2 Amino acid sequence of the different domains, others than the scFv, constituting the MSLN CARs according to the invention.
  • Table 3 Amino acid sequences of P4-R2, Meso1-R2, Mesd and MES02-R2 CARs.
  • Table 4 full polypeptide sequences of MSLN CARs, dnTGF ⁇ RII and MSLN epitope region
  • the signal transducing domain or intracellular signaling domain of a CAR is responsible for intracellular signaling following the binding of extracellular ligand binding domain to the target resulting in the activation of the immune cell and immune response.
  • the signal transducing domain is responsible for the activation of at least one of the normal effector functions of the immune cell in which the CAR is expressed.
  • the effector function of a T cell can be a cytolytic activity or helper activity including the secretion of cytokines.
  • the term “signal transducing domain” refers to the portion of a protein which transduces the effector signal function signal and directs the cell to perform a specialized function.
  • Preferred examples of signal transducing domain for use in a CAR can be the cytoplasmic sequences of the T cell receptor and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivate or variant of these sequences and any synthetic sequence that has the same functional capability.
  • Signal transduction domain comprises two distinct classes of cytoplasmic signaling sequence, those that initiate antigen-dependent primary activation, and those that act in an antigen-independent manner to provide a secondary or co-stimulatory signal.
  • Primary cytoplasmic signaling sequence can comprise signaling motifs which are known as immunoreceptor tyrosine-based activation motifs of ITAMs.
  • ITAMs are well defined signaling motifs found in the intracytoplasmic tail of a variety of receptors that serve as binding sites for syk/zap70 class tyrosine kinases.
  • Examples of ITAM used in the invention can include as non-limiting examples those derived from TCRzeta, FcRgamma, FcRbeta, FcRepsilon, CD3gamma, CD3delta, CD3epsilon, CD5, CD22, CD79a, CD79b and CD66d.
  • the signaling transducing domain of the CAR can comprise the CD3zeta signaling domain which has amino acid sequence with at least 70%, preferably at least 80%, more preferably at least 90 %, 95 % 97 % or 99 % sequence identity with amino acid sequence selected from the group consisting of (SEQ ID NO: 9).
  • the signal transduction domain of the CAR of the present invention comprises a co-stimulatory signal molecule.
  • a co-stimulatory molecule is a cell surface molecule other than an antigen receptor or their ligands that is required for an efficient immune response.
  • “Co-stimulatory ligand” refers to a molecule on an antigen presenting cell that specifically binds a cognate co-stimulatory molecule on a T-cell, thereby providing a signal which, in addition to the primary signal provided by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, mediates a T cell response, including, but not limited to, proliferation activation, differentiation and the like.
  • a co-stimulatory ligand can include but is not limited to CD7, B7-1 (CD80), B7-2 (CD86), PD-L1, PD-L2, 4-1 BBL, OX40L, inducible costimulatory ligand (ICOS-L), intercellular adhesion molecule (ICAM, CD30L, CD40, CD70, CD83, HLA-G, MICA, M1CB, HVEM, lymphotoxin beta receptor, 3/TR6, ILT3, ILT4, an agonist or antibody that binds Toll ligand receptor and a ligand that specifically binds with B7-H3.
  • a co-stimulatory ligand also encompasses, inter alia, an antibody that specifically binds with a co-stimulatory molecule present on a T cell, such as but not limited to, CD27, CD28, 4-1 BB, 0X40, CD30, CD40, PD-1 , ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LTGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83.
  • an antibody that specifically binds with a co-stimulatory molecule present on a T cell such as but not limited to, CD27, CD28, 4-1 BB, 0X40, CD30, CD40, PD-1 , ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LTGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83.
  • LFA-1 lymphocyte function-associated antigen-1
  • the signal transduction domain of the CAR of the present invention comprises a part of co-stimulatory signal molecule selected from the group consisting of fragment of 4-1 BB (GenBank: AAA53133.) and CD28 (NP_006130.1).
  • the signal transduction domain of the CAR of the present invention comprises amino acid sequence which comprises at least 70%, preferably at least 80%, more preferably at least 90 %, 95 % 97 % or 99 % sequence identity with either 4-1 BB or CD28.
  • the mesothelin specific chimeric antigen receptor as per the present invention preferably comprises a CD3 zeta signalling domain that has at least 80 % identity with SEQ ID NO.
  • a CAR according to the present invention is generally expressed on the surface membrane of the cell.
  • Such CAR further comprises a transmembrane domain.
  • the distinguishing features of appropriate transmembrane domains comprise the ability to be expressed at the surface of a cell, preferably in the present invention an immune cell, in particular lymphocyte cells or Natural killer (NK) cells, and to interact together for directing cellular response of immune cell against a predefined target cell.
  • the transmembrane domain can be derived either from a natural or from a synthetic source.
  • the transmembrane domain can be derived from any membrane-bound or transmembrane protein.
  • the transmembrane polypeptide can be a subunit of the T-cell receptor such as a, b, g or z, polypeptide constituting CD3 complex, IL2 receptor p55 (a chain), p75 (b chain) or g chain, subunit chain of Fc receptors, in particular Fey receptor III or CD proteins.
  • the transmembrane domain can be synthetic and can comprise predominantly hydrophobic residues such as leucine and valine.
  • said transmembrane domain is derived from the human CD8 alpha chain (e.g. NP_001139345.1)
  • the transmembrane domain can further comprise a hinge region between said extracellular ligand-binding domain and said transmembrane domain.
  • hinge region generally means any oligo- or polypeptide that functions to link the transmembrane domain to the extracellular ligand-binding domain.
  • hinge region are used to provide more flexibility and accessibility for the extracellular ligand-binding domain.
  • a hinge region may comprise up to 300 amino acids, preferably 10 to 100 amino acids and most preferably 25 to 50 amino acids.
  • Hinge region may be derived from all or part of naturally occurring molecules, such as from all or part of the extracellular region of CD8, CD4 or CD28, or from all or part of an antibody constant region.
  • the hinge region may be a synthetic sequence that corresponds to a naturally occurring hinge sequence, or may be an entirely synthetic hinge sequence.
  • said hinge domain comprises a part of human CD8 alpha chain, FcyRIIIa receptor or lgG1 respectively, or hinge polypeptides which display preferably at least 80%, more preferably at least 90 %, 95 % 97 % or 99 % sequence identity with these polypeptides.
  • mesothelin specific chimeric antigen receptor comprises a hinge between the extracellular ligand-binding domain and the transmembrane domain, said hinge being generally selected from CD8a hinge, lgG1 hinge and FcyRIIIa hinge or polypeptides sharing at least 80%, preferably at least 90%, more preferably at least 95%, and even more preferably at least 99% sequence identity with these polypeptides, in particular with SEQ ID NO:16 (CD8a).
  • a CAR according to the invention generally further comprises a transmembrane domain (TM) preferably selected from CD8a and 4-1 BB, more preferably from CD8a-TM or a polypeptide showing at least 80%, more preferably at least 90 %, 95 % 97 % or 99 % sequence identity with SEQ ID N0.17 (CD8a TM).
  • TM transmembrane domain
  • the mesothelin specific CAR according to the invention comprises a safety switch useful for sorting, purifying and/or depleting the engineered immune cells.
  • a safety switch useful for sorting, purifying and/or depleting the engineered immune cells.
  • Table 5 Examples of mAb-specific epitopes (and their corresponding mAbs), which can be inserted in the extracellular binding domain of the CAR of the invention.
  • a mesothelin specific CAR according to the invention preferably comprises a safety switch which comprises at least one exogenous mAb epitope listed in Table 5.
  • a mesothelin specific CAR according to the invention preferably comprises a safety switch which comprises the epitope CPYSNPSLC (SEQ ID NO:26) that is specifically bound by rituximab.
  • a mesothelin specific CAR comprises a safety switch referred to as “R2”, that has at least 90% identity with SEQ ID NO: 15.
  • a mesothelin specific CAR according to the invention generally also comprises a signal peptide to help its expression at the surface of the engineered cells.
  • the chimeric antigen receptor (CAR) generally form single-chain polypeptides, but may also be produced in multi chain formats as described for instance in WO2014039523.
  • preferred CARs according to the invention is MSLN- CAR-Meso1-R2 or MSLN-CAR-Meso1 , which has respectively at least 80%, preferably at least 90%, more preferably at least 95%, and even more preferably at least 99% overall amino acid sequence identity with SEQ ID NO:21 (Meso1-R2) or SEQ ID NO:22 (Mesol).
  • the CARs of the present invention are produced by assembling the different polynucleotides sequences encoding the successive fragments of the CAR polypeptide(s) into vectors for transfection and expression into immune cells as described in the art and as reviewed for instance by [Boyiadzis, M.M., et al. (2016) Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. / immunotherapy cancer 6, 137]
  • the present invention is drawn to the polynucleotides and vectors as well as any intermediary steps intervening in the process of manufacturing the immune cells referred herein.
  • vector is meant a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • a “vector” in the present invention includes, but is not limited to, a viral vector, a plasmid, a RNA vector or a linear or circular DNA or RNA molecule which may consists of a chromosomal, non-chromosomal, semi-synthetic or synthetic nucleic acids.
  • Preferred vectors are those capable of autonomous replhcation (episomal vector) and/or expression of nucleic acids to which they are linked (expression vectors). Large numbers of suitable vectors are known to those of skill in the art and commercially available.
  • Viral vectors include retrovirus, adenovirus, parvovirus (e. g. adeno-'associated viruses (AAV), coronavirus, negative strand RNA viruses such as ortho-myxovirus (e. g., influenza virus), rhabdovirus (e. g., rabies and vesicular stoma-'titis virus), para-myxovirus (e. g. measles and Sendai), positive strand RNA viruses such as piconnavirus and alphavirus, and double-stranded DNA viruses including adenovirus, herpesvirus (e. g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus), and poxvirus (e.
  • AAV adeno-'associated viruses
  • coronavirus negative strand RNA viruses
  • ortho-myxovirus e. g., influenza virus
  • rhabdovirus e. g., rabies and ves
  • viruses include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus, for example.
  • retroviruses include: avian leukosis-sarcoma, mammalian C-type, B-type viruses, D type viruses, HTLV-BLV group, lenthvirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields, et al. , Eds., Lippincott-Raven Publishers, Philadelphia, 1996).
  • the present invention provides inter alia an expression vector in the form of a lentiviral vector or an AAV vector comprising a polynucleotide sequence encoding a CAR as described herein.
  • Such lentiviral vector may comprise a polynucleotide sequence encoding a CAR according to the present invention operably linked to a promoter (such as the Spleen Focus Forming Virus promoter (SFFV)).
  • a promoter such as the Spleen Focus Forming Virus promoter (SFFV)
  • SFFV Spleen Focus Forming Virus promoter
  • operably linked it is meant a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a gene (such as a CAR encoding polynucleotide sequence) is "operably linked” to a promoter when its transcription is under the control of said promoter and this transcription results in the production of the product encoded by said gene.
  • the lentiviral vector of the present invention typically contains regulatory elements such as 5' and 3' long terminal repeat (LTR) sequences, but may also contain other structural and functional genetic elements that are primarily derived from a lentivirus. Such structural and functional genetic elements are well known in the art.
  • the lentiviral vector may, for example, contain the genes gag, pol and env. Preferably, however, the lentiviral vector of the present invention does not contain the genes gag, pol and env.
  • the lentiviral vector may include one or more (such as two or more) of a packaging signal (such as the packaging signal y), a primer binding site, a trans-activation-responsive region (TAR) and a rev-responsive element (RRE).
  • the 5' and 3' long terminal repeat (LTR) sequences typically flanking the lentiviral genome have promoter/enhancer activity and are essential for the correct expression of the full-length lentiviral vector transcript.
  • the LTRs usually include the repetitive sequence U3RU5 present at both the 5’- and 3’ ends of a double-stranded DNA molecule, which is a combination of 5’ R-U5 segment and the 3’ U3-R segment of the single-stranded RNA, wherein repetition R occurs at both termini of the RNA, while U5 (unique sequence 5) only occurs at the 5’ end of the RNA and U3 (unique sequence 3) only occurs at the 3’ end of the RNA.
  • Lentiviral vectors can been improved in their safety by removal of the U3 sequence, resulting in "self-inactivating" vectors that are entirely devoid of viral promoter and enhancer sequences originally present within the LTRs. Consequently, the vector is capable of infecting and then integrating into the host genome only once, and cannot be passed further, thereby increasing the safety of the use of the vector as a gene delivery vector.
  • the lentiviral vector is a self-inactivating (SIN) lentiviral vector.
  • the lentiviral vector contains a 3’ LTR in which the 3' LTR enhancer-promoter sequence (i.e. U3 sequence) has been modified (e.g., deleted).
  • the lentiviral vector comprises a polynucleotide sequence which comprises one or several of the following elements in a 5' to 3' order:
  • LTR long terminal repeat
  • the lentiviral vector can further comprise a polynucleotide sequence which comprises at least one of the following elements in a 5' to 3' order:
  • promoter such as the EF1 -alpha promoter
  • a CAR optionally comprising a safety switch, such as R2,
  • dnTGFbR a polynucleotide sequence encoding a or any further polypeptide to be co-expressed with the CAR, such as dnTGFbR;
  • LTR long terminal repeat
  • the lentiviral vector can comprise at least one of the following elements in a 5' to 3' order:
  • promoter such as the EF1 -alpha promoter
  • dnTGFbR a polynucleotide sequence encoding a or any further polypeptide to be co-expressed with the CAR, such as dnTGFbR;
  • a CAR optionally comprising a safety switch, such as R2,
  • LTR long terminal repeat
  • the resulting vector form a single transcription unit operably linked to the promoter of item (b) and are all transcribed under the control of said promoter.
  • AAV vectors especially vectors from the AAV6 family [Wang, J., et al. (2015) Homology- driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat Biotechnol 33, 1256-1263] are particularly useful to introduce the MSLN-CARs according to the present invention into the genome by using site-specific homologous recombination.
  • site specific homologous recombination is induced in immune cells by expressing rare-cutting endonucleases, such as TALEN, as already taught in EP3276000 and WO2018073391 with respect to other CARs for treating blood cancers.
  • CAR site-specific integration can have several benefits, such as a more stable integration, an integration that places the transgene under the transcription control of an endogenous promoter at a selected locus, an integration that can inactivate an endogenous locus.
  • an AAV vector comprising a polynucleotide sequence encoding a MSLN-CAR as previously specified and optionally another sequence encoding a cis-regulatory elements (e.g. 2A peptide cleavage site) or an internal ribosome entry site (IRES), allowing the co-expression of a third sequence encoding a product improving the therapeutic potency of the engineered immune cells.
  • a cis-regulatory elements e.g. 2A peptide cleavage site
  • IRS internal ribosome entry site
  • therapeutic properties encompasses the different ways such cells can be improved in the perspective of their use in therapeutic treatments.
  • the cells are genetically engineered to confer them a therapeutic advantage benefit (i.e. therapeutic potency) or to facilitate their use or their production.
  • the genetic engineering can concur to the effector cells having better survival, faster growth, shorter cell cycles, improved immune activity, be more functional, more differentiated, more specific with respect to their target cells, more sensitive or resistant to drugs, less sensitive to glucose deprivation, oxygen or amino acid depletion (i.e. resilient to tumor microenvironment).
  • Progenitor cells may be more productive, better tolerated by the recipient patient, more likely to produce cells that will differentiate in the desired effector cells.
  • Effector cells are the relatively short-lived activated cells that defend the body in an immune response.
  • Activated T cells which include cytotoxic T cells and helper T cells are preferred effector cells to carry out cell-mediated responses.
  • the category of effector T cell is a broad one that includes various T cell types that actively respond to a stimulus, such as co-stimulation. This includes helper, killer, regulatory, and potentially other T cell types.
  • immune cell is meant a cell of hematopoietic origin functionally involved in the initiation and/or execution of innate and/or adaptative immune response, such as typically CD3 or CD4 positive cells.
  • the immune cell according to the present invention may be a dendritic cell, killer dendritic cell, a mast cell, a NK-cell, a B-cell or a T-cell selected from the group consisting of inflammatory T-lymphocytes, cytotoxic T-lymphocytes, regulatory T-lymphocytes or helper T-lymphocytes.
  • primary cell or “primary cells” are intended cells taken directly from living tissue (e.g. biopsy material) and established for growth in vitro for a limited amount of time, meaning that they can undergo a limited number of population doublings. Primary cells are opposed to continuous tumorigenic or artificially immortalized cell lines.
  • Non-limiting examples of such cell lines are CHO-K1 cells; HEK293 cells; Caco2 cells; U2-OS cells; NIH 3T3 cells; NSO cells; SP2 cells; CHO-S cells; DG44 cells; K-562 cells, U-937 cells; MRC5 cells; IMR90 cells; Jurkat cells; HepG2 cells; HeLa cells; HT-1080 cells; HCT-116 cells; Hu-h7 cells; Huvec cells; Molt 4 cells.
  • Primary immune cells can be obtained from a number of non-limiting sources, including peripheral blood mononuclear cells (PBMC), bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and from tumors, such as tumor infiltrating lymphocytes.
  • PBMC peripheral blood mononuclear cells
  • said immune cell can be derived from a healthy donor, from a patient diagnosed with cancer or from a patient diagnosed with an infection.
  • said cell is part of a mixed population of immune cells which present different phenotypic characteristics, such as comprising CD4, CD8 and CD56 positive cells.
  • the immune cells derived from stem cells are also regarded as primary immune cells according to the present invention, in particular those deriving from induced pluripotent stem cells (iPS) [Yamanaka, K. et al. (2008). “Generation of Mouse Induced Pluripotent Stem Cells Without Viral Vectors”. Science. 322 (5903): 949-53]
  • Lentiviral expression of reprogramming factors has been used to induce multipotent cells from human peripheral blood cells [Staerk, J. et al. (2010). "Reprogramming of human peripheral blood cells to induced pluripotent stem cells”.
  • the immune cells are derived from human embryonic stem cells by techniques well known in the art that do not involve the destruction of human embryos [Chung et al. (2008) Human Embryonic Stem Cell lines generated without embryo destruction, Cell Stem Cell 2(2): 113-117]
  • Genetic engineering is meant any methods aiming to introduce, modify and/or withdraw genetic material from a cell.
  • gene editing is meant a genetic engineering allowing genetic material to be added, removed, or altered at specific locations (loci) in the genome, including punctual mutations. Gene editing generally involves sequence specific reagents.
  • sequence-specific reagent any active molecule that has the ability to specifically recognize a selected polynucleotide sequence at a genomic locus, referred to as “target sequence”, which is generally of at least 9 bp, more preferably of at least 10 bp and even more preferably of at least 12 pb in length, in view of modifying the expression of said genomic locus.
  • target sequence is generally of at least 9 bp, more preferably of at least 10 bp and even more preferably of at least 12 pb in length, in view of modifying the expression of said genomic locus.
  • Said expression can be modified by mutation, deletion or insertion into coding or regulatory polynucleotide sequences, by epigenetic change, such as by methylation or histone modification, or by interfering at the transcriptional level by interacting with transcription factors or polymerases.
  • sequence-specific reagents are endonucleases, RNA guides, RNAi, methylases, exonucleases, histone deacetylases, endonucleases, end-processing enzymes such as exonucleases, and more particularly cytidine deaminases such as those coupled with the CRISPR/cas9 system to perform base editing (i.e. nucleotide substitution) without necessarily resorting to cleavage by nucleases as described for instance by Hess, G.T. et al. [Methods and applications of CRISPR-mediated base editing in eukaryotic genomes (2017) Mol Cell. 68(1): 26-43.
  • said sequence-specific reagent is preferably a sequence-specific nuclease reagent, such as a RNA guide coupled with a guided endonuclease.
  • the present invention aims to improve the therapeutic potential of immune cells through gene editing techniques, especially by gene targeted integration.
  • gene targeting integration is meant any known site-specific methods allowing to insert, replace or correct a genomic coding sequence into a living cell.
  • said gene targeted integration involves homologous gene recombination at the locus of the targeted gene to result the insertion or replacement of at least one exogenous nucleotide, preferably a sequence of several nucleotides (i.e. polynucleotide), and more preferably a coding sequence.
  • exogenous nucleotide preferably a sequence of several nucleotides (i.e. polynucleotide), and more preferably a coding sequence.
  • DNA target a polynucleotide sequence that can be targeted and processed by a sequence -specific nuclease reagent according to the present invention.
  • These terms refer to a specific DNA location, preferably a genomic location in a cell, but also a portion of genetic material that can exist independently to the main body of genetic material such as plasmids, episomes, virus, transposons or in organelles such as mitochondria as non-limiting example.
  • RNA guided target sequences are those genome sequences that can hybridize the guide RNA which directs the RNA guided endonuclease to a desired locus.
  • “Rare-cutting endonucleases” are sequence-specific endonuclease reagents of choice, insofar as their recognition sequences generally range from 10 to 50 successive base pairs, preferably from 12 to 30 bp, and more preferably from 14 to 20 bp.
  • said endonuclease reagent is a nucleic acid encoding an “engineered” or “programmable” rare-cutting endonuclease, such as a homing endonuclease as described for instance by Arnould S., et al. [W02004067736], a zinc finger nuclease (ZFN) as described, for instance, by Urnov F., et al. [Highly efficient endogenous human gene correction using designed zinc-finger nucleases (2005) Nature 435:646-651], a TALE-Nuclease as described, for instance, by Mussolino et al.
  • an “engineered” or “programmable” rare-cutting endonuclease such as a homing endonuclease as described for instance by Arnould S., et al. [W02004067736], a zinc finger nuclease (ZFN) as described, for instance, by Urnov F
  • the endonuclease reagent is a RNA-guide to be used in conjunction with a RNA guided endonuclease, such as Cas9 or Cpf1, as per, inter alia, the teaching by Doudna, J., and Chapentier, E., [The new frontier of genome engineering with CRISPR-Cas9 (2014) Science 346 (6213):1077], which is incorporated herein by reference.
  • a RNA guided endonuclease such as Cas9 or Cpf1
  • the endonuclease reagent is transiently expressed into the cells, meaning that said reagent is not supposed to integrate into the genome or persist over a long period of time, such as be the case of RNA, more particularly mRNA, proteins or complexes mixing proteins and nucleic acids (eg: Ribonucleoproteins).
  • An endonuclease under mRNA form is preferably synthetized with a cap to enhance its stability according to techniques well known in the art, as described, for instance, by Kore A.L., et al. [Locked nucleic acid (LNA)-modified dinucleotide mRNA cap analogue: synthesis, enzymatic incorporation, and utilization (2009 ) J Am Chem Soc. 131(18):6364-5]
  • LNA locked nucleic acid
  • electroporation steps that are used to transfect primary immune cells, such as PBMCs are typically performed in closed chambers comprising parallel plate electrodes producing a pulse electric field between said parallel plate electrodes greater than 100 volts/cm and less than 5,000 volts/cm, substantially uniform throughout the treatment volume such as described in W02004083379, which is incorporated by reference, especially from page 23, line 25 to page 29, line 11.
  • One such electroporation chamber preferably has a geometric factor (cm -1 ) defined by the quotient of the electrode gap squared (cm2) divided by the chamber volume (cm 3 ), wherein the geometric factor is less than or equal to 0.1 cm -1 , wherein the suspension of the cells and the sequence-specific reagent is in a medium which is adjusted such that the medium has conductivity in a range spanning 0.01 to 1.0 milliSiemens.
  • the suspension of cells undergoes one or more pulsed electric fields.
  • the treatment volume of the suspension is scalable, and the time of treatment of the cells in the chamber is substantially uniform.
  • TALE-nuclease Due to their higher specificity, TALE-nuclease have proven to be particularly appropriate sequence specific nuclease reagents for therapeutic applications, especially under heterodimeric forms - i.e. working by pairs with a “right” monomer (also referred to as “5”’ or “forward”) and left” monomer (also referred to as “3”” or “reverse”) as reported for instance by Mussolino et a/.
  • TALEN facilitate targeted genome editing in human cells with high specificity and low cytotoxicity (2014) Nucl. Acids Res. 42(10): 6762-6773]
  • sequence specific reagent is preferably under the form of nucleic acids, such as under DNA or RNA form encoding a rare cutting endonuclease a subunit thereof, but they can also be part of conjugates involving polynucleotide(s) and polypeptide(s) such as so-called “ribonucleoproteins”.
  • conjugates can be formed with reagents as Cas9 or Cpf1 (RNA-guided endonucleases) as respectively described by Zetsche, B. et al. [Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System (2015) Cell 163(3): 759-771] and by Gao F. et al. [DNA-guided genome editing using the Natronobacterium gregoryi Argonaute (2016) Nature Biotech], which involve RNA or DNA guides that can be complexed with their respective nucleases.
  • Exogenous sequence refers to any nucleotide or nucleic acid sequence that was not initially present at the selected locus. This sequence may be homologous to, or a copy of, a genomic sequence, or be a foreign sequence introduced into the cell. By opposition “endogenous sequence” means a cell genomic sequence initially present at a locus. The exogenous sequence preferably codes for a polypeptide which expression confers a therapeutic advantage over sister cells that have not integrated this exogenous sequence at the locus. An endogenous sequence that is gene edited by the insertion of a nucleotide or polynucleotide as per the method of the present invention, in order to express a different polypeptide is broadly referred to as an exogenous coding sequence.
  • the present invention concurs to develop methods for producing therapeutic cells by proceeding with one or several of the following steps:
  • sequence specific-reagent such as rare- cutting endonuclease to induce a modification (mutations or coding sequence insertion) at an endogenous gene locus;
  • the immune cells originate from a patient or a compatible donor, in which the MSLN CAR is expressed in view of performing so called “autologous” infusion of the engineered immune cells. They can also be derived from stem cells, such as iPS cells, originating from such patient or compatible donor or from tumor infiltrating lymphocytes (TILL).
  • stem cells such as iPS cells, originating from such patient or compatible donor or from tumor infiltrating lymphocytes (TILL).
  • the method aims to provide “off the shelf” compositions of immune cells, said immune cells being engineered for allogeneic therapeutic treatments.
  • allogeneic is meant that the cells originate from a donor, is produced or differentiated from stem cells in view of being infused into patients having a different haplotype.
  • Such immune cells are generally engineered to be less alloreactive and/or become more persistent with respect to their patient host. More specifically the present methods comprise the steps of reducing or inactivating TCR expression into T-cells, or stem cells to be derived into T-cells. This can be obtained by different sequence specific-reagents, such as by gene silencing or gene editing techniques (nuclease, base editing, RNAi).
  • TALE-nucleases TALEN ®
  • the present invention provides with a method to engineer an immune cell, wherein at least one gene encoding TCRalpha or TCRbeta is inactivated in said immune cell, preferably by expression of a rare-cutting endonuclease, whereas an exogenous polynucleotide encoding MSLN-CAR is introduced into the genome of said cell for stable expression.
  • said exogenous sequence is integrated at said locus encoding TCRalpha or TCRbeta, more preferably under transcriptional control of an endogenous promoter of TCRalpha or TCRbeta.
  • the engineered immune cell can be further modified to confer resistance to at least one immune suppressive drug, such as by inactivating CD52 the target of anti-CD52 antibody (e.g.:alemtuzumab), which has been previously described with respect to the treatment of blood cancers for instance in WO2013176915.
  • an immune suppressive drug such as by inactivating CD52 the target of anti-CD52 antibody (e.g.:alemtuzumab), which has been previously described with respect to the treatment of blood cancers for instance in WO2013176915.
  • one major aspect of the present invention is the use of genetically engineered lymphocytes made resistant to lymphodepletion regimen for the treatment of solid tumors.
  • the present invention provides engineered lymphocytes endowed with chimeric antigen receptors directed against solid tumors, especially against mesothelin positive cells, for their use in solid tumor cancer treatments in combination with, or preceded by, a lymphodepletion treatment step.
  • Such lymphodepletion regimen can comprise anti-CD52 reagents, such as Alemtuzumab, or purine analogues, as those used for treating blood cancers.
  • anti-CD52 reagents such as Alemtuzumab, or purine analogues, as those used for treating blood cancers.
  • the engineered lymphocytes endowed with MSLN-CAR described herein are made resistant to such lymphodepleting regimen by inhibiting or disrupting the expression of the molecules that are targeted by the lymphodepletion reagents, like for instance the antigen CD52 in the case of Alemtuzumab.
  • the engineered immune cell can be further modified to confer resistance to and/or a chemotherapy drug, in particular a purine analogue drug, for example by inactivating DCK as described in WO201575195.
  • Such regimen can comprise antibodies targeting antigens present at the surface of immune cells, such as CD52, CD3, CD4, CD8, CD45, or other specific markers, but also less specific drugs such as purine analogues (ex: fludarabine and/or chlorofarabine) and glucocorticoids.
  • One aspect of the invention is to make the engineered lymphocytes resistant to such regimen by inactivating or reducing the expression of the genes that encode at least one molecular target of these lymphodepletion reagents, for instance the gene DCK that metabolizes purine analogues or the genes encoding glucocorticoid receptors (GR).
  • the genes that encode at least one molecular target of these lymphodepletion reagents for instance the gene DCK that metabolizes purine analogues or the genes encoding glucocorticoid receptors (GR).
  • the present invention is therefore more particularly focused on CAR positive cells, which expression of TCR, CD52 and/or DCK and/or GR is reduced, inactivated or deficient to make them less alloreactive and resistant to lymphodepletion regimen, for their allogeneic use in solid cancer treatments.
  • the engineered immune cell can be further modified to improve its persistence or its lifespan into the patient, in particular inactivating a gene encoding MHC-I component(s) such as HLA or b2hi, such as described in W02015136001 or by Liu, X. et al. [CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells (2017) Cell Res 27:154-157]
  • MHC-I component(s) such as HLA or b2hi, such as described in W02015136001 or by Liu, X. et al. [CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells (2017) Cell Res 27:154-157]
  • the engineered immune cell is mutated to improve its CAR-dependent immune activation, in particular to reduce or suppress the expression of immune checkpoint proteins and/or their receptors thereof, such as PD1 or CTLA4 as described in WO2014184744.
  • the engineered immune cell can be further modified to obtain co-expression in said cell of another exogenous genetic sequence selected from one encoding:
  • - NK cell inhibitor such as HLAG, HLAE or ULBP1;
  • - CRS inhibitor such as is a mutated IL6Ra, sGP130 or IL18-BP;
  • DHFR Dihydrofolate reductase
  • IMPDH2 inosine monophosphate dehydrogenase 2
  • MGMT calcineurin or methylguanine transferase
  • mTORmut conferring drug resistance
  • cytokine such as IL-2, IL-12 and IL-15;
  • Chemokine receptors such as CCR2, CXCR2, or CXCR4;
  • TAM Tumor Associated Macrophages
  • the present application claims immune cells co-expressing in engineered immune cells at least one exogenous sequence encoding MSLN-CAR as described herein, with another exogenous sequence encoding a human polypeptide selected in the above list for the purpose of producing therapeutic compositions against solid tumors. Combination of expression of MSLN-CAR and disruption of TGFbRII signalling pathway in the therapeutic engineered immune cells
  • the present invention more particularly combines the expression of an exogenous sequence encoding MSLN-CAR as previously described, with another exogenous sequence encoding an inhibitor of a TGFbeta receptor, especially an inhibitor of TGF ⁇ RII (Uniprot - P37173).
  • TGFbeta receptors have been described as having preponderant roles in tumor microenvironment [Papageorgis, P. et al. (2015). Role of TGF ⁇ in regulation of the tumor microenvironment and drug delivery (Review). International Journal of Oncology, 46, 933- 943]
  • TGFbeta receptors Although, the exact role of TGFbeta receptors in tumorogenesis remains controversial, the inventors have found that co-expressing mesothelin-specific chimeric antigen receptor (CAR) with another exogenous genetic sequence encoding an inhibitor of TGFBRII signalling and/or inactivating or reducing TGFbeta receptor signalling by using a sequence-specific reagent, was leading to an improved therapeutic potency of the engineered immune cells. In particular, the inventors have used two different approaches to impair TGFbRII signalling pathway, which may be combined together:
  • TGFbRII an inactive ligand of TGFbRII, such as a dominant negative TGFbRII (SEQ ID NO:26), as described by Hiramatsu, K., et al. [Expression of dominant negative TGF- b receptors inhibits cartilage formation in conditional transgenic mice (2011) J. Bone. Miner. Metab. 29: 493] or a similar inactive form of TGF ⁇ EII having at least 80%, preferably at least 90%, more preferably at least 95% identity with the polypeptide sequence SEQ ID NO:26. and/or
  • TGF ⁇ EII RNA-guided endonuclease
  • a rare cutting endonuclease such as a TALE-nuclease or RNA-guided endonuclease (e.g.:Cas9 or Cpf1).
  • An anti- TGF ⁇ RII IgGi monoclonal antibody that inhibits receptor-mediated signaling activation such as LY3022859 [Tolcher, A.W. et al. (2017) A phase 1 study of anti-TGF ⁇ receptor type-ll monoclonal antibody LY3022859 in patients with advanced solid tumors Cancer Chemother Pharmacol.79(4): 673-680] can also be used to inhibit TGFbeta receptor signalling in combination of the CAR as per the present invention.
  • TALE-nucleases which are particularly specific to a selection of target sequences within the TGF ⁇ EII gene. These TALE- nucleases have displayed highest TGF ⁇ EII knock-out efficiency with very little off-target cleavage resulting into large populations of viable engineered cells, sufficient for dosing several patients. These preferred TALE-nuclease and their corresponding target sequences are listed in Table 6.
  • Table 6 TALE-nuclease target sequences for TGF ⁇ RII gene.
  • RNA guides devise also been designed to inactivate TGF ⁇ RII gene by using Cas9 nuclease reagent. Their corresponding respective target sequences are disclosed in Table 7.
  • Table 7 CRISPR target sequences for TGFbRII gene.
  • the present invention thus encompasses the use of a TALE-nuclease or RNA-guided endonuclease designed to bind any of the target sequences SEQ ID NO:X to Y referred to in table 5 or 6 for the inactivation or reducing of expression of TGF ⁇ RII for the production of therapeutic immune cells within the teaching of the present specification.
  • the present invention also pertains to engineered immune cells comprising an exogenous polynucleotide encoding a nuclease, such as one referred to before, to inactivate or reduce the expression of its endogenous TGF ⁇ RII gene.
  • the present application thus reports engineered immune cells, especially CAR immune cells, into which an exogenous sequence encoding an inhibitor of TGFbeta receptor has been introduced, more particularly a sequence encoding a dominant negative TGFbeta receptor.
  • Such cells are more particularly dedicated to the treatment of solid tumors, especially MSLN positive tumors.
  • vectors especially viral vectors, such as lentiviral vectors or AAV vectors as described in the art, comprising at least a polynucleotide sequence encoding a dominant negative TGFbRII, and optionally, a mesothelin-specific chimeric antigen receptor.
  • said vectors comprise a first polynucleotide sequence encoding said dominant negative TGFbRII, a second polynucleotide sequence encoding 2A self-cleaving peptide and a third one encoding said mesothelin-specific chimeric antigen receptor.
  • Targeted insertion into immune cells can be significantly improved by using AAV vectors, especially vectors from the AAV6 family or chimeric vectors AAV2/6 previously described by Sharma A., et al. [Transduction efficiency of AAV 2/6, 2/8 and 2/9 vectors for delivering genes in human corneal fibroblasts. (2010) Brain Research Bulletin. 81 (2-3): 273- 278]
  • One aspect of the present invention is thus the transduction of AAV vectors comprising MSLN-CAR coding sequence in human primary immune cells, in conjunction with the expression of sequence-specific endonuclease reagents, such as TALE endonucleases, to increase gene integration at the loci previously cited.
  • sequence specific endonuclease reagents can be introduced into the cells by transfection, more preferably by electroporation of mRNA encoding said sequence specific endonuclease reagents.
  • the obtained insertion of the exogenous nucleic acid sequence may result into the introduction of genetic material, correction or replacement of the endogenous sequence, more preferably “in frame” with respect to the endogenous gene sequences at that locus.
  • from 10 5 to 10 7 preferably from 10 6 to 10 7 , more preferably about 5.10 6 viral genomes viral genomes are transduced per cell.
  • the cells can be treated with proteasome inhibitors, such as Bortezomib or HDAC inhibitors to further help homologous recombination.
  • proteasome inhibitors such as Bortezomib or HDAC inhibitors
  • the AAV vector used in the method can comprise an exogenous coding sequence that is promoter less, said coding sequence being any of those referred to in this specification.
  • the present invention also provides with an efficient method for obtaining primary immune cells, which can be gene edited in various gene loci more particularly involved into host-graft interaction and recognition. Other loci may also be edited in view of improving the activity, the survival or the life-time of the engineered primary cells, especially primary T cells.
  • Figure 2 maps the main cell functions that can be modified by gene editing according to the present invention to improve the efficiency of the engineered immune cells. Any gene inactivation listed under each function can be combined with another to obtain a synergistic effect on the overall therapeutic potency of the immune cells.
  • the present invention provides more particularly with combinations of genetic modifications (genotypes) into immune cells prompt to improve immune cells potency against solid tumor, especially against MSLN positive malignant cells, such as:
  • CAR positive cells in solid cancer treatments, which are made resistant to lymphodepleting agents, so that they can be combined with or preceded by lymphodepletion regimen for their use in allogeneic settings.
  • Such cells preferably display the following genotypes:
  • the above preferred genotypes can be obtained by gene targeting integration, preferably at the PD1, TCR (TCRalpha and/or TCRbeta) or TGF ⁇ RII loci, but also at further selected loci as described here after.
  • Gene targeting integration is meant any known site-specific methods allowing to insert, replace or correct a genomic sequence into a living cell.
  • Gene targeted integration usually involves the mechanisms of homologous gene recombination or NHEJ (Non homologous Ends Joining), which are enhanced by endonuclease sequence specific reagents, to result into insertion or replacement of at least one exogenous nucleotide, preferably a sequence of several nucleotides (i.e. polynucleotide), and more preferably a coding sequence at a predefined locus.
  • NHEJ Non homologous Ends Joining
  • the method according to the invention comprises the steps of introducing into an immune cell a mutation or polynucleotide coding sequence at an endogenous locus selected from: a) polynucleotide sequence(s), which expression is(are) involved into reduction of glycolysis and calcium signaling in response to a low glucose condition, such as SERCA3 to increase calcium signaling, miR101 and mir26Ato increase glycolysis, BCAT to mobilize glycolytic reserves; and/or b) polynucleotide sequence(s), which expression up regulate(s) immune checkpoint proteins (e.g.TIM3, CEACAM, LAG3, TIGIT), such as IL27RA, STAT1, STAT3; and/or c) polynucleotide sequence(s), which expression mediate(s) interaction with HLA-G, such as ILT
  • Said transgene or exogenous polynucleotide sequence is preferably inserted so that its expression is placed under transcriptional control of at least one endogenous promoter present at one of said locus.
  • Targeting one locus as referred to above by performing gene integration is beneficial to further improve the potency of the therapeutic immune cells of the invention.
  • the exogenous sequence that is integrated into the immune cells genomic locus encodes a molecule that confers resistance of said immune cells to a drug.
  • DHFR dihydrofolate reductase
  • folate analogs such as methotrexate
  • variants of inosine monophosphate dehydrogenase 2 (IMPDH2) conferring resistance to IMPDH inhibitors
  • MPA mycophenolic acid
  • MMF prodrug mycophenolate mofetil
  • variants of calcineurin or methylguanine transferase (MGMT) conferring resistance to calcineurin inhibitor such as FK506 and/or CsA
  • variants of mTOR such as mTORmut conferring resistance to rapamycin
  • variants of Lck such as Lckmut conferring resistance to Imatinib and Gleevec.
  • drug is used herein as referring to a compound or a derivative thereof, preferably a standard chemotherapy agent that is generally used for interacting with a cancer cell, thereby reducing the proliferative or living status of the cell.
  • chemotherapeutic agents include, but are not limited to, alkylating agents (e.g., cyclophosphamide, ifosamide), metabolic antagonists (e.g., purine nucleoside antimetabolite such as clofarabine, fludarabine or 2’-deoxyadenosine, methotrexate (MTX), 5-fluorouracil or derivatives thereof), antitumor antibiotics (e.g., mitomycin, adriamycin), plant-derived antitumor agents (e.g., vincristine, vindesine, Taxol), cisplatin, carboplatin, etoposide, and the like.
  • alkylating agents e.g., cyclophosphamide,
  • Such agents may further include, but are not limited to, the anti-cancer agents TRIMETHOTRIXATETM (TMTX), TEMOZOLOMIDETM, RALTRITREXEDTM , S-(4-Nitrobenzyl)- 6-thioinosine (NBMPR),6-benzyguanidine (6-BG), bis-chloronitrosourea (BCNU) and CAMPTOTHECINTM, or a therapeutic derivative of any thereof.
  • TTTX TRIMETHOTRIXATETM
  • TEMOZOLOMIDETM TEMOZOLOMIDETM
  • RALTRITREXEDTM S-(4-Nitrobenzyl)- 6-thioinosine
  • 6-BG 6-benzyguanidine
  • BCNU bis-chloronitrosourea
  • CAMPTOTHECINTM CAMPTOTHECINTM
  • an immune cell is made "resistant or tolerant" to a drug when said cell, or population of cells is modified so that it can proliferate, at least in-vitro, in a culture medium containing half maximal inhibitory concentration (IC50) of said drug (said IC50 being determined with respect to an unmodified cell(s) or population of cells).
  • IC50 half maximal inhibitory concentration
  • said drug resistance can be conferred to the immune cells by the expression of at least one “drug resistance coding sequence”.
  • Said drug resistance coding sequence refers to a nucleic acid sequence that confers "resistance" to an agent, such as one of the chemotherapeutic agents referred to above.
  • a drug resistance coding sequence of the invention can encode resistance to anti-metabolite, methotrexate, vinblastine, cisplatin, alkylating agents, anthracyclines, cytotoxic antibiotics, anti-immunophilins, their analogs or derivatives, and the like (Takebe, N., S. C. Zhao, et al.
  • DHFR Dihydrofolate reductase
  • MTX methotrexate
  • the drug resistance coding sequence according to the present invention can be a nucleic acid sequence encoding a mutant form of human wild type DHFR (GenBank: AAH71996.1), which comprises at least one mutation conferring resistance to an anti-folate treatment, such as methotrexate.
  • mutant form of DHFR comprises at least one mutated amino acid at position G15, L22, F31 or F34, preferably at positions L22 or F31 (Schweitzer et al. (1990) "Di hydrofolate reductase as a therapeutic target" Faseb 4(8): 2441-52; International application W094/24277; and US patent US 6,642,043).
  • said DHFR mutant form comprises two mutated amino acids at position L22 and F31. Correspondence of amino acid positions described herein is frequently expressed in terms of the positions of the amino acids of the form of wild-type DHFR polypeptide.
  • the serine residue at position 15 is preferably replaced with a tryptophan residue.
  • the leucine residue at position 22 is preferably replaced with an amino acid which will disrupt binding of the mutant DHFR to antifolates, preferably with uncharged amino acid residues such as phenylalanine or tyrosine.
  • the phenylalanine residue at positions 31 or 34 is preferably replaced with a small hydrophilic amino acid such as alanine, serine or glycine.
  • Another example of drug resistance coding sequence can also be a mutant or modified form of ionisine-5’- monophosphate dehydrogenase II (IMPDH2), a rate-limiting enzyme in the de novo synthesis of guanosine nucleotides.
  • the mutant or modified form of IMPDH2 is a IMPDH inhibitor resistance gene.
  • IMPDH inhibitors can be mycophenolic acid (MPA) or its prodrug mycophenolate mofetil (MMF).
  • MMF prodrug mycophenolate mofetil
  • the mutant IMPDH2 can comprises at least one, preferably two mutations in the MAP binding site of the wild type human IMPDH2 (Genebank: NP_000875.2) leading to a significantly increased resistance to IMPDH inhibitor.
  • Mutations in these variants are preferably at positions T333 and/or S351 (Yam, P., M. Jensen, et a/. (2006) "Ex vivo selection and expansion of cells based on expression of a mutated inosine monophosphate dehydrogenase 2 after HIV vector transduction: effects on lymphocytes, monocytes, and CD34+ stem cells” Mol. Ther. 14(2): 236-44)(Jonnalagadda, M., et a/. (2013) "Engineering human T cells for resistance to methotrexate and mycophenolate mofetil as an in vivo cell selection strategy.” PLoS One 8(6): e65519).
  • Calcineurin is an ubiquitously expressed serine/threonine protein phosphatase that is involved in many biological processes and which is central to T-cell activation. Calcineurin is a heterodimer composed of a catalytic subunit (CnA; three isoforms) and a regulatory subunit (CnB; two isoforms). After engagement of the T-cell receptor, calcineurin dephosphorylates the transcription factor NFAT, allowing it to translocate to the nucleus and active key target gene such as IL2.
  • CnA catalytic subunit
  • CnB regulatory subunit
  • said mutant form can comprise at least one mutated amino acid of the wild type calcineurin heterodimer a at positions: V314, Y341, M347, T351 , W352, L354, K360, preferably double mutations at positions T351 and L354 or V314 and Y341.
  • the valine residue at position 341 can be replaced with a lysine or an arginine residue
  • the tyrosine residue at position 341 can be replaced with a phenylalanine residue
  • the methionine at position 347 can be replaced with the glutamic acid, arginine or tryptophane residue
  • the threonine at position 351 can be replaced with the glutamic acid residue
  • the tryptophane residue at position 352 can be replaced with a cysteine, glutamic acid or alanine residue
  • the serine at position 353 can be replaced with the histidine or asparagines residue
  • the leucine at position 354 can be replaced with an alanine residue
  • the lysine at position 360 can be replaced with an alanine or phenylalanine residue.
  • said mutant form can comprise at least one mutated amino acid of the wild type calcineurin heterodimer b at positions: V120, N123, L124 or K125, preferably double mutations at positions L124 and K125.
  • the valine at position 120 can be replaced with a serine, an aspartic acid, phenylalanine or leucine residue;
  • the asparagines at position 123 can be replaced with a tryptophan, lysine, phenylalanine, arginine, histidine or serine;
  • the leucine at position 124 can be replaced with a threonine residue;
  • the lysine at position 125 can be replaced with an alanine, a glutamic acid, tryptophan, or two residues such as leucine-arginine or isoleucine-glutamic acid can be added after the lysine at position 125 in the amino acid sequence.
  • Correspondence of amino acid positions described herein is frequently expressed in terms of
  • AGT is a DNA repair protein that confers resistance to the cytotoxic effects of alkylating agents, such as nitrosoureas and temozolomide (TMZ).
  • TMZ nitrosoureas and temozolomide
  • 6-benzylguanine (6-BG) is an inhibitor of AGT that potentiates nitrosourea toxicity and is co-administered with TMZ to potentiate the cytotoxic effects of this agent.
  • AGT mutant form can comprise a mutated amino acid of the wild type AGT position P140.
  • said proline at position 140 is replaced with a lysine residue.
  • Another drug resistance coding sequence can be multidrug resistance protein (MDR1) gene.
  • MDR1 multidrug resistance protein
  • This gene encodes a membrane glycoprotein, known as P-glycoprotein (P-GP) involved in the transport of metabolic byproducts across the cell membrane.
  • P-GP P-glycoprotein
  • the P-Gp protein displays broad specificity towards several structurally unrelated chemotherapy agents.
  • drug resistance can be conferred to cells by the expression of nucleic acid sequence that encodes MDR-1 (Genebank NP_000918).
  • Another drug resistance coding sequence can contribute to the production of cytotoxic antibiotics, such as those from ble or mcrA genes. Ectopic expression of ble gene or mcrA in an immune cell gives a selective advantage when exposed to the respective chemotherapeutic agents bleomycine and mitomycin C (Belcourt, M.F. (1999) “Mitomycin resistance in mammalian cells expressing the bacterial mitomycin C resistance protein MCRA”. PNAS. 96(18):10489-94).
  • Another drug resistance coding sequence can come from genes encoded mutated version of drug targets, such as mutated variants of mTOR (mTOR mut) conferring resistance to rapamycin such as described by Lorenz M.C. et al. (1995) “TOR Mutations Confer Rapamycin Resistance by Preventing Interaction with FKBP12-Rapamycin” The Journal of Biological Chemistry 270, 27531-27537, or certain mutated variants of Lck (Lckmut) conferring resistance to Gleevec as described by Lee K.C. et al. (2010) “Lck is a key target of imatinib and dasatinib in T-cell activation”, Leukemia, 24: 896-900.
  • mTOR mut mutated variants of mTOR
  • Lckmut Lckmut
  • the genetic modification step of the method can comprise a step of introduction into cells of an exogeneous nucleic acid comprising at least a sequence encoding the drug resistance coding sequence and a portion of an endogenous gene such that homologous recombination occurs between the endogenous gene and the exogeneous nucleic acid.
  • said endogenous gene can be the wild type “drug resistance” gene, such that after homologous recombination, the wild type gene is replaced by the mutant form of the gene which confers resistance to the drug.
  • the exogenous sequence that is integrated into the immune cells genomic locus encodes a molecule that enhances persistence of the immune cells, especially in-vivo persistence in a tumor environment.
  • enhancing persistence is meant extending the survival of the immune cells in terms of life span, especially once the engineered immune cells are injected into the patient. For instance, persistence is enhanced, if the mean survival of the modified cells is significantly longer than that of non-modified cells, by at least 10%, preferably 20%, more preferably 30%, even more preferably 50%.
  • a various panel of such polypeptides in particular antagonists of immune checkpoints, immunosuppressive peptides derived from viral envelope or NKG2D ligand can enhance persistence and/or an engraftment of allogeneic immune cells into patients.
  • the immunosuppressive polypeptide to be encoded by said exogenous coding sequence is a ligand of Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4 also known as CD152, GenBank accession number AF414120.1).
  • Said ligand polypeptide is preferably an anti-CTLA-4 immunoglobulin, such as CTLA-4a Ig and CTLA-4b Ig or a functional variant thereof.
  • the immunosuppressive polypeptide to be encoded by said exogenous coding sequence is an antagonist of PD1 , such as PD-L1 (other names: CD274, Programmed cell death 1 ligand; ref. UniProt for the human polypeptide sequence Q9NZQ7), which encodes a type I transmembrane protein of 290 amino acids consisting of a Ig V-like domain, a Ig C-like domain, a hydrophobic transmembrane domain and a cytoplasmic tail of 30 amino acids.
  • PD1 such as PD-L1 (other names: CD274, Programmed cell death 1 ligand; ref. UniProt for the human polypeptide sequence Q9NZQ7), which encodes a type I transmembrane protein of 290 amino acids consisting of a Ig V-like domain, a Ig C-like domain, a hydrophobic transmembrane domain and a cytoplasmic tail of 30 amino acids.
  • Such membrane-bound form of PD-L1 ligand is meant in the present invention under a native form (wild-type) or under a truncated form such as, for instance, by removing the intracellular domain, or with one or more mutation(s) (Wang S et al. , 2003, J Exp Med. 2003; 197(9): 1083-1091).
  • PD1 is not considered as being a membrane-bound form of PD-L1 ligand according to the present invention.
  • said immunosuppressive polypeptide is under a secreted form.
  • Such recombinant secreted PD-L1 may be generated by fusing the extracellular domain of PD-L1 to the Fc portion of an immunoglobulin (Haile ST et a/., 2014, Cancer Immunol. Res. 2(7): 610-615; Song MY et al., 2015, Gut. 64(2):260-71).
  • This recombinant PD-L1 can neutralize PD-1 and abrogate PD-1-mediated T-cell inhibition.
  • PD-L1 ligand may be co-expressed with CTLA4 Ig for an even enhanced persistence of both.
  • the exogenous sequence encodes a non-human MHC homolog, especially a viral MHC homolog, or a chimeric ⁇ 2m polypeptide such as described by Margalit A. etal. (2003) polypeptides expressed in T cells convert MHC class I peptide ligands into T cell activation receptors: a potential tool for specific targeting of pathogenic CD8+ T cells” Int. Immunol. 15 (11): 1379-1387.
  • the exogenous sequence encodes NKG2D ligand.
  • Some viruses such as cytomegaloviruses have acquired mechanisms to avoid NK cell mediate immune surveillance and interfere with the NKG2D pathway by secreting a protein able to bind NKG2D ligands and prevent their surface expression (Welte, S.A et al. (2003) “Selective intracellular retention of virally induced NKG2D ligands by the human cytomegalovirus UL16 glycoprotein”. Eur. J. Immunol., 33, 194-203).
  • NKG2D ligands such as ULBP2, MICB or MICA (Salih HR, Antropius H, Gieseke F, Lutz SZ, Kanz L, et a/. (2003) Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. Blood 102: 1389-1396).
  • the exogenous sequence encodes a cytokine receptor, such as an IL-12 receptor.
  • IL-12 is a well known activator of immune cells activation (Curtis J.H. (2008) “I L-12 Produced by Dendritic Cells Augments CD8+ T Cell Activation through the Production of the Chemokines CCL1 and CCL171”. The Journal of Immunology. 181 (12): 8576-8584.
  • the exogenous sequence encodes an antibody that is directed against inhibitory peptides or proteins.
  • Said antibody is preferably be secreted under soluble form by the immune cells.
  • Nanobodies from shark and camels are advantageous in this respect, as they are structured as single chain antibodies (Muyldermans S. (2013) “Nanobodies: Natural Single-Domain Antibodies” Annual Review of Biochemistry 82: 775- 797). Same are also deemed more easily to fuse with secretion signal polypeptides and with soluble hydrophilic domains.
  • the exogenous sequence that is integrated into the immune cells genomic locus encodes a molecule that enhances the therapeutic activity of the immune cells.
  • enhancing the therapeutic activity is meant that the immune cells, or population of cells, engineered according to the present invention, become more aggressive than non- engineered cells or population of cells with respect to a selected type of target cells.
  • Said target cells consists of a defined type of cells, or population of cells, preferably characterized by common surface marker(s).
  • therapeutic potential reflects the therapeutic activity, as measured through in-vitro experiments. In general sensitive cancer cell lines, such as Daudi cells, are used to assess whether the immune cells are more or less active towards said cells by performing cell lysis or growth reduction measurements. This can also be assessed by measuring levels of degranulation of immune cells or chemokines and cytokines production.
  • Experiments can also be performed in mice with injection of tumor cells, and by monitoring the resulting tumor expansion. Enhancement of activity is deemed significant when the number of developing cells in these experiments is reduced by the immune cells by more than 10%, preferably more than 20%, more preferably more than 30 %, even more preferably by more than 50 %.
  • said exogenous sequence encodes a chemokine or a cytokine, such as IL-12. It is particularly advantageous to express IL-12 as this cytokine is extensively referred to in the literature as promoting immune cell activation (Colombo M.P. et al. (2002) “lnterleukin-12 in anti-tumor immunity and immunotherapy” Cytokine Growth Factor Rev. 13(2): 155-68).
  • the exogenous coding sequence encodes or promote secreted factors that act on other populations of immune cells, such as T- regulatory cells, to alleviate their inhibitory effect on said immune cells.
  • said exogenous sequence encodes an inhibitor of regulatory T-cell activity is a polypeptide inhibitor of forkhead/winged helix transcription factor 3 (FoxP3), and more preferably is a cell-penetrating peptide inhibitor of FoxP3, such as that referred as P60 (Casares N. et al. (2010) “A peptide inhibitor of FoxP3 impairs regulatory T cell activity and improves vaccine efficacy in mice.” J Immunol 185(9):5150-9).
  • FoxP3 forkhead/winged helix transcription factor 3
  • inhibitor of regulatory T-cells activity is meant a molecule or precursor of said molecule secreted by the T-cells and which allow T-cells to escape the down regulation activity exercised by the regulatory T-cells thereon.
  • inhibitor of regulatory T-cell activity has the effect of reducing FoxP3 transcriptional activity in said cells.
  • said exogenous sequence encodes a secreted inhibitor of Tumor Associated Macrophages (TAM), such as a CCR2/CCL2 neutralization agent.
  • TAM Tumor-associated macrophages
  • CCR2/CCL2 neutralization agent Tumor-associated macrophages
  • TAMs are critical modulators of the tumor microenvironment.
  • Clinicopathological studies have suggested that TAM accumulation in tumors correlates with a poor clinical outcome. Consistent with that evidence, experimental and animal studies have supported the notion that TAMs can provide a favorable microenvironment to promote tumor development and progression. (Theerawut C. et al. (2014) “Tumor-Associated Macrophages as Major Players in the Tumor Microenvironment” Cancers (Basel) 6(3): 1670-1690).
  • Chemokine ligand 2 (CCL2), also called monocyte chemoattractant protein 1 (MCP1 - NCBI NP_002973.1), is a small cytokine that belongs to the CC chemokine family, secreted by macrophages, that produces chemoattraction on monocytes, lymphocytes and basophils.
  • CCR2 C-C chemokine receptor type 2 - NCBI NP_001116513.2
  • CCL2 C-C chemokine receptor type 2 - NCBI NP_001116513.2
  • the coding sequence which is inserted at said locus generally encodes polypeptide(s) improving the therapeutic potential of the engineered immune cells
  • the inserted sequence can also be a nucleic acid able to direct or repress expression of other genes, such as interference RNAs or guide-RNAs.
  • the polypeptides encoded by the inserted sequence may act directly or indirectly, such as signal transducers or transcriptional regulators.
  • the present invention is also drawn to the variety of engineered immune cells obtainable according to one of the method described herein, under isolated form, or as part of populations of cells.
  • the engineered cells are primary immune cells, such as NK cells or T-cells, which are generally part of populations of cells that may involve different types of cells.
  • primary immune cells such as NK cells or T-cells
  • NK cells or T-cells are generally part of populations of cells that may involve different types of cells.
  • the present invention encompasses immune cells comprising any combinations of the different exogenous coding sequences and gene inactivation, which have been respectively and independently described above. Among these combinations are particularly preferred those combining the expression of a CAR under the transcriptional control of an endogenous promoter that is active during immune cell activation, in particular one promoter present at one TCR locus, in particular a TCRalpha promoter.
  • Another preferred combination is the insertion of an exogenous sequence encoding a CAR or one of its constituents under the transcription control of the hypoxia-inducible factor 1 gene promoter (Uniprot: Q16665).
  • the invention is also drawn to a pharmaceutical composition comprising an engineered primary immune cell or immune cell population as previously described for the treatment of infection or cancer, and to a method for treating a patient in need thereof, wherein said method comprises: preparing a population of engineered primary immune cells according to the method of the invention as previously described; optionally, purifying or sorting said engineered primary immune cells; activating said population of engineered primary immune cells upon or after infusion of said cells into said patient.
  • the immune cells according to the present invention can be activated or expanded, even if they can activate or proliferate independently of antigen binding mechanisms.
  • T-cells in particular, can be activated and expanded using methods as described, for example, in U.S. Patents 6,352,694; 6,534,055; 6,905,680;
  • T-cells can be expanded in vitro or in vivo. T cells are generally expanded by contact with an agent that stimulates a CD3 TCR complex and a co-stimulatory molecule on the surface of the T-cells to create an activation signal for the T-cell.
  • chemicals such as calcium ionophore A23187, phorbol 12-myristate 13-acetate (PMA), or mitogenic lectins like phytohemagglutinin (PHA) can be used to create an activation signal for the T-cell.
  • PMA phorbol 12-myristate 13-acetate
  • PHA phytohemagglutinin
  • T cell populations may be stimulated in vitro such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore.
  • a protein kinase C activator e.g., bryostatin
  • a ligand that binds the accessory molecule is used for co-stimulation of an accessory molecule on the surface of the T cells.
  • a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells.
  • Conditions appropriate for T cell culture include an appropriate media (e.g., Minimal Essential Media or RPMI Media 1640 or, X-vivo 5, (Lonza)) that may contain factors necessary for proliferation and viability, including serum (e.g., fetal bovine or human serum), interleukin-2 (IL-2), insulin, IFN-g, IL-4, IL-7, GM-CSF, IL-10, IL-2, IL-15, TGFp, and TNF- or any other additives for the growth of cells known to the skilled artisan.
  • Other additives for the growth of cells include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetyl-cysteine and 2-mercaptoethanoi.
  • Media can include RPMI 1640, A1M-V, DMEM, MEM, a-MEM, F-12, X- Vivo 1 , and X-Vivo 20, Optimizer, with added amino acids, sodium pyruvate, and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokine(s) sufficient for the growth and expansion of T cells.
  • Antibiotics e.g., penicillin and streptomycin, are included only in experimental cultures, not in cultures of cells that are to be infused into a subject.
  • the target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37° C) and atmosphere (e.g., air plus 5% C02). T cells that have been exposed to varied stimulation times may exhibit different characteristics.
  • said cells can be expanded by co-culturing with tissue or cells. Said cells can also be expanded in vivo, for example in the subject’s blood after administrating said cell into the subject.
  • the method of the present invention described above allows producing engineered primary immune cells within a limited time frame of about 15 to 30 days, preferably between 15 and 20 days, and most preferably between 18 and 20 days so that they keep their full immune therapeutic potential, especially with respect to their cytotoxic activity.
  • These cells form a population of cells, which preferably originate from a single donor or patient. These populations of cells can be expanded under closed culture recipients to comply with highest manufacturing practices requirements and can be frozen prior to infusion into a patient, thereby providing “off the shelf” or “ready to use” therapeutic compositions.
  • PBMC comprises several types of cells: granulocytes, monocytes and lymphocytes, among which from 30 to 60 % of T-cells, which generally represents between 10 8 to 10 9 of primary T-cells from one donor.
  • the method of the present invention generally ends up with a population of engineered cells that reaches generally more than about 10 8 T- cells, more generally more than about 10 9 T-cells, even more generally more than about 10 10 T-cells, and usually more than 10 11 T-cells.
  • the invention is thus more particularly drawn to a therapeutically effective population of primary immune cells, wherein at least 30 %, preferably 50 %, more preferably 80 % of the cells in said population have been modified according to any one the methods described herein.
  • more than 50% of the immune cells comprised in said population are TCR negative T-cells. According to a more preferred aspect of the invention, more than 50% of the immune cells comprised in said population are CAR positive T-cells.
  • compositions or populations of cells can therefore be used as medicaments; especially for treating cancer, particularly for the treatment of lymphoma, but also for solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas such as lung, breast, colon, prostate or ovary tumors in a patient in need thereof.
  • solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas such as lung, breast, colon, prostate or ovary tumors in a patient in need thereof.
  • the invention is more particularly drawn to populations of primary TCR negative T-cells originating from a single donor, wherein at least 20 %, preferably 30 %, more preferably 50 % of the cells in said population have been modified using sequence-specific reagents in at least two, preferably three different loci.
  • the present invention relies on methods for treating patients in need thereof, said method comprising at least one of the following steps:
  • said populations of cells mainly comprises CD4 and CD8 positive immune cells, such as T-cells, which can undergo robust in vivo T cell expansion and can persist for an extended amount of time in-vitro and in-vivo.
  • the treatments involving the engineered primary immune cells according to the present invention can be ameliorating, curative or prophylactic. It may be either part of an autologous immunotherapy or part of an allogenic immunotherapy treatment.
  • said isolated cell according to the invention or cell line derived from said isolated cell can be used for the treatment of solid tumors, in particular solid tumors, such as typically: oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, lung cancer, thymic carcinoma, mesothelioma, ovarian cancer and/or endometrial cancer.
  • solid tumors such as typically: oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, lung cancer, thymic carcinoma, mesothelioma, ovarian cancer and/or endometrial cancer.
  • the treatment with the engineered immune cells according to the invention may be in combination with one or more therapies against cancer selected from the group of antibodies therapy, chemotherapy, cytokines therapy, dendritic cell therapy, gene therapy, hormone therapy, laser light therapy and radiation therapy.
  • said treatment can be administrated into patients undergoing an immunosuppressive treatment.
  • the present invention preferably relies on cells or population of cells, which have been made resistant to at least one immunosuppressive agent due to the inactivation of a gene encoding a receptor for such immunosuppressive agent.
  • the immunosuppressive treatment should help the selection and expansion of the T-cells according to the invention within the patient.
  • the administration of the cells or population of cells according to the present invention may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation.
  • the compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous or intralymphatic injection, or intraperitoneally.
  • the cell compositions of the present invention are preferably administered by intravenous injection.
  • the administration of the cells or population of cells can consist of the administration of 10 4 - 10 9 cells per kg body weight, preferably 10 5 to 10 6 cells/kg body weight including all integer values of cell numbers within those ranges.
  • the present invention thus can provide more than 10, generally more than 50, more generally more than 100 and usually more than 1000 doses comprising between 10 6 to 10 8 gene edited cells originating from a single donor’s or patient’s sampling.
  • the cells or population of cells can be administrated in one or more doses.
  • said effective amount of cells are administrated as a single dose.
  • said effective amount of cells are administrated as more than one dose over a period time. Timing of administration is within the judgment of managing physician and depends on the clinical condition of the patient.
  • the cells or population of cells may be obtained from any source, such as a blood bank or a donor. While individual needs vary, determination of optimal ranges of effective amounts of a given cell type for a particular disease or conditions within the skill of the art.
  • An effective amount means an amount which provides a therapeutic or prophylactic benefit.
  • the dosage administrated will be dependent upon the age, health and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment and the nature of the effect desired.
  • said effective amount of cells or composition comprising those cells are administrated parenterally.
  • Said administration can be an intravenous administration.
  • Said administration can be directly done by injection within a tumor.
  • cells are administered to a patient in conjunction with (e.g., before, simultaneously or following) any number of relevant treatment modalities, including but not limited to treatment with agents such as antiviral therapy, cidofovir and interleukin-2, Cytarabine (also known as ARA-C) or nataliziimab treatment for MS patients or efaliztimab treatment for psoriasis patients or other treatments for PML patients.
  • agents such as antiviral therapy, cidofovir and interleukin-2, Cytarabine (also known as ARA-C) or nataliziimab treatment for MS patients or efaliztimab treatment for psoriasis patients or other treatments for PML patients.
  • the T cells of the invention may be used in combination with chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludaribine, cyclosporin, FK506, rapamycin, mycoplienolic acid, steroids, FR901228, cytokines, and irradiation.
  • immunosuppressive agents such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies
  • immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies
  • cytoxin fludaribine
  • cyclosporin FK506, rapamycin
  • mycoplienolic acid steroids
  • steroids FR901228
  • cytokines irradiation
  • the cell compositions of the present invention are administered to a patient in conjunction with (e.g., before, simultaneously or following) bone marrow transplantation, T cell ablative therapy using either chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH,
  • the cell compositions of the present invention are administered following B-cell ablative therapy such as agents that react with CD20, e.g., Rituxan.
  • subjects may undergo standard treatment with high dose chemotherapy followed by peripheral blood stem cell transplantation.
  • subjects receive an infusion of the expanded immune cells of the present invention.
  • expanded cells are administered before or following surgery.
  • the present invention is also particularly drawn to a general method of treating solid tumor(s) in a patient, comprising the steps of immunodepleting said patient with a lymphodepletion regimen and infusing genetically engineered lymphocytes made resistant to the lymphodepletion agent used in the lymphodepletion regimen and specifically targeting said solid tumor(s).
  • Such genetically engineered lymphocytes are preferably CAR positive T-cells, more preferably endowed with a MSLN-CAR as described herein.
  • the lymphodepletion regimen preferably comprises an antibody directed against an antigen present at the surface of immune cells, such as CD52, CD3, CD4, CD8, CD45, or other specific markers, or being drugs such as purine analogues (ex: fludarabine and/or chlorofarabine) and glucocorticoids.
  • an antigen present at the surface of immune cells such as CD52, CD3, CD4, CD8, CD45, or other specific markers, or being drugs such as purine analogues (ex: fludarabine and/or chlorofarabine) and glucocorticoids.
  • the method comprises submitting the patient to a lymphodepletion regimen comprising an antibody directed against CD52, and administrating an engineered CAR T-cell endowed with a MSLN-CAR, which expression of CD52 is reduced, deficient or inactivated,.
  • the lymphodepleting treatment can comprise an anti-CD52 antibody, such as alemtuzumab, alone or in combination.
  • the lymphodepletion regimen may for instance combine cyclophosphamide, typically for 1 to 3 days, fludarabine for 1 to 5 days, and alemtuzumab from 1 to 5 days.
  • the lymphodepletion regimen can comprise cyclophosphamide between 50 and 70 mg/kg/day, fludarabine between 20 and 40 mg/m2/day, and alemtuzumab 0,1 to 0,5 mg/kg/day alone or in combination.
  • the present invention provides with the combined use of a composition for lymphodepleting a patient affected by a solid tumor, said composition comprising an anti-CD52 antibody, and a population of engineered lymphocytes targeting MSLN that are not sensitive to said antibody, such population preferably comprising cells that express MSLN-CAR and have impaired CD52 expression.
  • allele(s) of the CD52 gene has been preferably inactivated by a rare-cutting endonuclease, such as a TALE-nuclease or a RNA-guided endonuclease as previously described.
  • the present invention also provides with a medical kit comprising said lymphodepleting composition and said population of engineered cells resistant thereto for its use in solid tumors cancer treatment.
  • cytolytic activity or “cytotoxic activity” or “cytotoxicity” is meant the percentage of cell lysis of target cells conferred by an immune cell.
  • STA specific target antigen
  • STA-negative cells 2.10 4 specific target antigen (STA)-positive or STA-negative cells are seeded in 0.1ml per well in a 96 well plate. The day after the plating, the STA-positive and the STA-negative cells are labeled with CellTrace CFSE and co-cultured with 4 x 10 5 T cells for 4 hours. The cells are then harvested, stained with a fixable viability dye (eBioscience) and analyzed using the MACSQuant flow cytometer (Miltenyi).
  • a fixable viability dye eBioscience
  • STA-positive and STA-negative cells are respectively labeled with CellTrace CFSE and CellTrace Violet.
  • About 2 x 10 4 ROR1 -positive cells are co cultured with 2 x 10 4 STA-negative cells with 4 x 10 5 T cells in 0.1ml per well in a 96-well plate. After a 4 hour incubation, the cells are harvested and stained with a fixable viability dye (eBioscience) and analyzed using the MACSQuant flow cytometer (Miltenyi).
  • the percentage of specific lysis can be calculated using the following formula:
  • % cell lysis of target cells conferred by the engineered immune cells is increased by at least 10%, such as at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 100% or more, compared to the % cell lysis of target cells conferred by the immune cell not being engineered.
  • identity refers to sequence identity between two nucleic acid molecules or polypeptides. Identity can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base, then the molecules are identical at that position. A degree of similarity or identity between nucleic acid or amino acid sequences is a function of the number of identical or matching nucleotides at positions shared by the nucleic acid sequences.
  • Various alignment algorithms and/or programs may be used to calculate the identity between two sequences, including FASTA, or BLAST which are available as a part of the GCG sequence analysis package (University of Wisconsin, Madison, Wis.), and can be used with, e.g., default setting.
  • polypeptides having at least 70%, 85%, 90%, 95%, 98% or 99% identity to specific polypeptides described herein and preferably exhibiting substantially the same functions, as well as polynucleotide encoding such polypeptides, are contemplated.
  • subject or “patient” as used herein generally refers to mammalians, preferably to primates and more preferably to humans.
  • MSLN Mesothelin
  • GPI glycophosphatidylinsositol
  • Mesothelin is reported to be highly expressed in several types of malignant tumors, such as malignant mesothelioma, ovarian cancer, pancreatic adenocarcinoma, and lung adenocarcinoma (Morello et al., 2016; O’Hara et al., 2016). In some cases, mesothelin expression has been associated with increased tumor aggressiveness and poor clinical outcome. Description of the MSLN-specific CARs used in the study
  • Three second-generation CARs composed of the respective scFvs P4, meso1 and MES02, comprising CD8a hinge/transmembrane domain, and the 4-1 BB and activation domains, were generated and screened for chimeric antigen receptor (CAR) expression and anti-tumor activity in vitro, through the activity of primary mesoCAR + T-cells against target cell lines expressing different levels of Mesothelin (MSLN).
  • a suicide switch “R2” was introduced in the CAR architecture.
  • the “R2” polypeptide which includes two CD20 mimotopes, was located between the scFv and the hinge to confer sensitivity to anti-CD20 therapeutic antibodies, such as rituximab, as previously described in W02016120216.
  • CARs were screened for expression in primary T-cells from PBMC and assayed for their anti tumor activity against 3 target cell lines expressing different levels of MSLN, respectively:
  • PBMC Peripheral Blood Mononuclear Cells
  • PBMC peripheral blood mononuclear cells
  • X-vivo 15 medium supplemented with 5% AB serum, 350UI/ml IL2 and MACS GMP T Cell TransAct (60mI per million of CD3+ cells).
  • the cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells and rl_V vectors bearing the polynucleotide sequences encoding the anti MSLN CARs were resuspended in X-vivo 15 medium supplemented with 5% AB serum and 350UI/ml IL2, and seeded over retronectin coated plates.
  • T cells were washed and resuspended in X-vivo 15 medium-supplemented with 5% AB serum, 350UI/ml IL2. The cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were co-electroporated with mRNA encoding the right and the left arms of respectively the TRAC TALEN and CD52 TALEN. as previously reported [Poirot eta/. (2013) Blood. 122 (21): 1661] to efficiently inactivate TCRa and CD52 gene and prevent TCRc ⁇ expression at the surface of the primary T-cells.
  • TALEN is the registered name for the TALE- nucleases designed by Cellectis (8, rue de la Croix Jarry, 75013 Paris, France). The genomic target sequences for these TALE nucleases are indicated in Table 8 below. Transfection was performed using the AgilePulse technology. The cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were washed and resuspended in X-vivo 15 medium-supplemented with 5% AB serum, 350UI/ml IL2. The cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were expanded in GRex devices.
  • GRex 6 multi-well cell culture plates were used, half the culture media was removed at days 11 and 15 and replaced with fresh medium containing IL2, and fresh IL2 was added at day 13.
  • fresh IL2 was added at days 11, 13 and 15 without any medium change.
  • the cell cultures were incubated at 37°C under 5% C02.
  • the totality of the UCART cells was cryopreserved for later used in in vitro and in vivo assays.
  • P4-R2, Meso1-R2 and MES02-R2 CARs were used to generate three MSLN-specific UCART cell products. The different UCART cell products were then evaluated in vitro.
  • CAR and TCRap expression at the surface of UCART cells, as well as CD4 and CD8 expression at the surface of the CAR+ fraction of UCART cells were analyzed by flow cytometry.
  • CAR surface expression was evaluated using either His-tagged recombinant human mesothelin protein, or biotinylated protein L, that all recognize the scFv portion of the CARs or biotinylated rituximab that recognizes the R2 suicide switch portion of the CARs.
  • TCRap receptor surface expression was evaluated using PE-vio770 conjugated anti-TCRap antibody.
  • CD4and CD8 surface expression were evaluated using FITC-conjugated CD4 and BV510-conjugated CD8 antibodies.
  • CAR surface expression was evaluated by flow cytometry using either His-tagged recombinant human mesothelin protein that recognizes the scFv portion of the CARs, or biotinylated rituximab that recognizes the R2 suicide switch portion of the CARs.
  • UCART cells modified with P4-R2 and Meso1-R2 CARs produced repectively more than 40000pg/ml and 50000pg/ml of IFNg upon coculture with a MSLN+ cell line and less than 1500 and 200 pg/ml of IFNg respectively upon coculture with a MSLN- cell line.
  • UCART cells modified with Meso1-R2 CAR produced similar level of IFNg than UCART cells modified with P4-R2 CARs upon coculture with a MSLN+ cell line
  • the Meso1-R2 CAR endowed cells produced extremely low level of IFNg upon culture with a MSLN- cell line.
  • UCART cells modified with MES02-R2 CAR produced no more than 15000pg/ml of IFNg upon coculture with a MSLN+ cell line.
  • the capacity of UCART cells to perform serial killing of HPAC cells was then evaluated over a period of 15 days including 6 rounds of exposure to MSLN+ (HPAC) cells at ratios 1:2 and 1:8.
  • CART cells modified with P4-R2, Meso1-R2 and MES02-R2 CARs displayed similar level of killing activity after a single round of exposure to HPAC cells.
  • T-cells endowed with Meso1-R2 and P4-R2 displayed much higher serial killing activity than the CART cells endowed with MES02-R2 CARs, while CART cells modified with Meso1-R2 showed more sustained activity than those modified with P4-R2
  • TCR ⁇ + was an efficient step to remove non engineered cells and select [CAR] + [TCR] UCART cells: 84% of non-engineered T-cells were TCRap+ as compared to 8% for the T- cells knocked-out for the TRAC gene and 0.2% for the T-cells knocked-out for the TRAC gene and depleted in TCRap+ cells.
  • Inactivation of CD52 genes was assessed by incubating the engineered Cells after 7 days of culture in 50 ⁇ g/ml anti-CD52 monoclonal antibody (or rat IgG as control) with or without 30% rabbit complement (Cedarlane). After 2 hours of incubation at 37°C, the cells were labeled with a fluorochrome-conjugated anti-CD52 antibody together with a fluorescent viability dye (eBioscience) and analyzed by flow cytometry to measure the frequency of CD52-positive and CD52-negative cells among live cells. On another hand the cells were cultured with the antibody to select resistant
  • UCART cells modified with P4-R2, Meso1-R2 and MES02-R2 CARs were co-cultured for two days with HPAC cells, exposed for two hours to medium, rituximab (RTX), baby rabbit complement (BRC) or a mixture of rituximab and baby rabbit complement, and analyzed by flow cytometry to monitor the percentage of CAR+ cells.
  • RTX rituximab
  • BRC baby rabbit complement
  • a mixture of rituximab and baby rabbit complement a mixture of rituximab and baby rabbit complement
  • TGFbRII TGFbRII
  • Activated T cells were electroporated with 10 ⁇ g of mRNA encoding the right and the left arms of two TALENs targeting TGFbRII gene (SEC ID NO:155 (pCLS32939) and SEC ID NO:156 (PCLS32940) or SEC ID NO:157 (pCLS32967) and SEC ID NO:158 (pCLS32968)).
  • SEC ID NO:155 pCLS32939
  • SEC ID NO:156 PCLS32940
  • SEC ID NO:157 pCLS32967
  • SEC ID NO:158 pCLS32968
  • PCR products were analyzed by deepsequencing and results show that transfection with TALEN encoded by pCLS32939 and pCLS32940 or with TALEN encoded by pCLS32967 and pCLS32968 showed 96.62% and 97.28% of gene editions (i.e. insertions and/or deletions) respectively, demonstrating the high efficiency of TGFbRII KO.
  • HPAC tumor cells injected sub cutaneously (SC) in NSG mice was selected as the animal/tumor model to be used to evaluate the anti-tumor activity in vivo of T-cells expressing the 3 selected mesoCAR constructs.
  • meso1-R2 was below in terms of level of expression at the cell surface, cytotoxicity and IFNy secretion, this was kept in the study and compared to P4-R2 and MES02-R2 CARs
  • mice Human T-cells expressing the mesoCAR candidates and the P4-CAR were then evaluated for their anti-tumor activity in vivo. Briefly, NSG mice were engrafted with a MSLN + cell line (HPAC cells, SC injection) and then treated with mesoCAR + T-cells (IV injection, 3 doses). mesoCAR + T-cells activity was assessed by the monitoring of tumor growth.
  • MSLN + cell line HPAC cells, SC injection
  • mesoCAR + T-cells IV injection, 3 doses.
  • the 3 mesoCAR + T-cells evaluated showed anti-tumor activity in vivo against HPAC tumor cells with different level of activity.
  • T-cells expressing the MES02-R2 CAR showed less activity compared to the other mesoCAR + T-cells evaluated.
  • the animal model selected is the highly immunodeficient NSG mice strain strain from the Jackson laboratory) as it allows the engraftment of both human MSLN + tumor cells and human CAR T- cells.
  • HeLa (ATCC ® CCL-2), an epithelial cervix adenocarcinoma, and HPAC (ATCC ® CRL-2119), an epithelial pancreatic adenocarcinoma
  • the aim of this first study was to evaluate the tumor take and tumor growth parameters of HeLa and HPAC cells after subcutaneous (SC) injection of NSG mice (6-8 weeks old).
  • mice were randomized according to their individual body weight into 4 groups of 6 mice each and received a SC injection of HeLa (1x10 6 or 10x10 6 cells/mouse) or HPAC cells (2x10 6 or 10x10 6 cells/mouse). Guantity of tumor cells were chosen according to the literature [Abate-Daga, D., et al. (2014). A Novel Chimeric Antigen Receptor Against Prostate Stem Cell Antigen Mediates Tumor Destruction in a Humanized Mouse Model of Pancreatic. Cancer. Hum. Gene Ther, Arjomandnejad et a/.(2014) Hela cell line xenograft tumor as a suitable cervical cancer model: Growth kinetic characterization and immunohistochemis-try array.
  • mice Body weight, viability and behavior were monitored every day. The tumor volume was measured three times a week. Surviving mice were terminated at Day 61 (end of study). Necropsy (macroscopic examination) was performed on all terminated animal in the study, and, if possible, on all euthanized moribund or found dead animal.
  • HPAC tumor grew faster than Hela tumor.
  • Mean tumor volume V (500 mm 3 ) for mice injected with 1x10 6 and 10x10 6 HeLa cells was respectively 519 mm 3 and 498 mm 3 with a mean time to reach of 55 days and 49 days.
  • Mean tumor volume V (500mm 3 ) for mice injected with 2x10 6 and 10x10 6 HPAC cells was respectively 557mm 3 and 500mm 3 with a mean time to reach of 24 days and 23 days.
  • mice were randomized according to their individual body weight into 2 groups of 3 mice each and received a SC injection of HeLa (10x10 6 cells/mouse) or HPAC cells (2x10 6 cells/mouse). Body weight, viability and behavior were monitored every day. The tumor volume was measured three times a week. Tumors were collected when tumor volume reached 300-500mm 3 . Tumor samples were analyzed by immunohistochemical (IHC) analysis of mesothelin expression. Last mouse was sacrificed at Day 63 (end of study).
  • IHC immunohistochemical
  • HPAC tumors have grown faster than Hela tumors.
  • the mean tumor volume of 300 mm 3 for mice injected with HeLa cells or HPAC cells was respectively reached at 40 days and 28 days.
  • mesothelin was checked using IHC on tumors collected on the mice. Both tumors expressed mesothelin.
  • HPAC cells (2x10 6 cells, SC injected) to evaluate the anti-tumor activity of mesoCAR + T-cells.
  • HPAC cells expressing firefly luciferase and GFP were generated by Cellectis and the tumor take and tumor growth of HPAC-luc-GFP cells in NSG mice was evaluated as previously described.
  • mice received a SC injection of HPAC-luc-GFP cells (2x10 6 cells/mouse). Body weight, viability and behavior were monitored every day. The tumor volume was measured three times a week and bioluminescence imaging was performed on Day 7, 14 and 24. Viability and behavior were monitored every day.
  • the aim of this study was to compare the anti-tumor activity of the 3 UCARTmeso candidates (P4-R2, MES02-R2 and meso1-R2) in NSG mice bearing subcutaneous HPAC tumors using the treatment conditions defined using the pilot study.
  • 3 doses of CAR + T-cells were evaluated (1 , 3 and 10x10 6 CAR positive cells / mouse).
  • UCARTmeso and control T-cells were produced with PBMCs from the same donor.
  • UCARTmeso and controls T-cells were not purified for TCRc ⁇ -negative cells. Characteristics of the T-cells used are in Table 9.
  • Table 9 Characteristics of the T-cells used in the study.
  • mice received a subcutaneous injection of HPAC tumor cells (2x10 6 cells/mouse) at Day -7.
  • 80 tumor bearing mice were randomized according to the tumor volume into 16 groups of 5 mice.
  • Human T-cells (UCARTmeso cells and KO TRAC/NT cells in control) were injected on Day 0 (for groups 1 to 15) or on Day 12 (for group 16).
  • the total number of cells injected was defined according to the percentage of CAR positive cells in the batch in order to inject the indicated number of CAR positive cells per mouse (1, 3 or 10x10 6 CAR positive cells).
  • UCARTmeso candidates CARs P4-R2, Meso1-R2 and MES02-R2
  • All UCARTmeso cells showed an anti-tumor activity, although with different level of activity between the different CAR T-cells.
  • Meso1-R2 CAR T-cells candidate is displaying the highest in-vivo activity at the 3 evaluated doses .
  • mice were challenged with 2 UCARTmeso candidates (P4-R2, MES01-R2) that are also expressing dnTGFBRII.
  • P4-R2, MES01-R2 2 UCARTmeso candidates
  • mice per group received a SC injection of HPAC cells (2x10 6 cells/mouse).
  • HPAC cells 2x10 6 cells/mouse.
  • the tumor volume was measured three times a week. Viability and behavior were monitored every day.
  • Table 10 Description of the six types of genetically modified T cells generated for the study.
  • PBMC Peripheral Blood Mononuclear Cells
  • PBMC peripheral blood mononuclear cells
  • T cells and rl_V vectors bearing the polynucleotide sequences encoding the different anti mesothelin CARs P4-CAR (SEQ ID NO:161) and MES01 CAR (SEQ ID NO:22) with or without the dnTGFBRII gene (SEQ ID NO: 24) were resuspended in X-vivo 15 medium supplemented with 5% AB serum and 350UI/ml IL2, and seeded over retronectin coated plates. The plates were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were washed and resuspended in X-vivo 15 medium-supplemented with 5% AB serum, 350UI/ml IL2. The cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were electroporated with mRNA encoding the right and the left arms of the TRAC TALEN and with or without mRNA encoding the right and left arms of TGFBRII TALEN (SEQ ID NO:157 and SEQ ID NO:158). Transfection was performed using the AgilePulse technology. The cells were then transferred to an incubator set at 37°C, 5% CQ2.
  • T cells were washed and resuspended in X-vivo 15 medium-supplemented with 5% AB serum, 350UI/ml IL2. The cells were then transferred to an incubator set at 37°C, 5% C02.
  • T cells were expanded in GRex devices. During the expansion period, the cell cultures were incubated at 37°C under 5% C02 and culture medium was changed from time to time.
  • the totality of the UCART cells was cryopreserved for later used in in vitro and in vivo assays.
  • the UCART were analyzed by flow cytometry.
  • CAR surface expression was evaluated using biotinylated recombinant mesothelin protein, that recognize the scFv portion of the CARs and PE-conjugated streptavidin.
  • the dnTGFBRII was evaluated using an anti-TGFBRII (from Abeam) and an APC-conjugated anti mouse IgG antibody.
  • CD4 and CD8 surface expression were evaluated using FITC-conjugated CD4 and BV510-conjugated CD8 antibodies.
  • the sternness of the UCART cells in CAR+CD4+ or CAR+ CD8+ positive cells where analyzed using anti-CD62L conjugated to PECy7 and anti-CD45RA coupled to APC.
  • the percentage of CD8+ positive cells was varying between 27 and 35% when the UCART cells expressed the P4 CAR construct.
  • the percentage CD8+ cells (among CAR positive cells) was varying from 36 to 51% (Figure 20B).
  • T cells genetically modified as indicated in Table 9 were mixed with H226-Luc/GFP cells at 1:3, 1:1, 3:1 , 10:1 effector-to-target (E:T) ratios. Cocultures were then incubated overnight at 37°C and bioluminescence was measured to quantify the lysis of H226 cells.
  • these genetically modified T cells were resuspended in culture medium and seeded at a density of 200000 cells / well in 96 well untreated or previously coated with 75ng / well of His-tagged recombinant mesothelin protein plates. Following a 24 hours incubation period, cell supernatants were collected and analyzed by ELISA to quantify the production of IFNg.
  • the UCART expressing MES01 could induce higher cytotoxicity at the lowest doses (1 :3 and 1 :1 ratio) than the UCART expressing P4.
  • Figure 23A demonstrates that the recombinant mesothelin protein was able to induce IFNg secretion in all the UCAR T produced. This production was varying from 40,000 up to 90,000 pg/ml and no obvious impact of the CAR construct or the TGFB pathway inhibition could be observed. However, when analyzing the IFNg secretion in absence of recombinant mesothelin ( Figure 23B), surprisingly the MES01 CAR construct was producing less IFNg than the P4 construct. This result suggests that UCART cells expressing MES01 CAR constructs are less stimulated in absence of antigen, which means that MES01 CAR has reduced “auto activation”. This is an important property in therapeutic settings because “auto activation” tends to exhaust CAR T-cells.
  • the genetically modified T cells as described in Table 9 were exposed to TGFb for one hour and were analyzed by flow cytometry to evaluate the fraction of pSMAD2/3 positive vs pSMAD2/3 negative cells among CAR positive cells.
  • the produced UCART cells were exposed to recombinant mesothelin protein in presence or not of TGFb and were counted 7 days later to assess proliferation.
  • Figure 24A shows that without inhibition of TGFb pathway, more than 95% of the CAR positive fraction of UCART cells were pSMAD2/3 positive.
  • FIG. 24B shows that TGFb is able to inhibit the antigen-mediated proliferation of UCART expressing P4 or MES01 construct to the same extent. Importantly TGFB pathway inhibition by either KO or dnTGFBRII overexpression are able to decrease or even abolish such inhibition.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Biotechnology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
EP20845393.6A 2019-12-23 2020-12-22 Neue mesothelin-spezifische chimäre antigen-rezeptoren (car) für die krebsimmuntherapie bei soliden tumoren Pending EP4081537A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA201970835 2019-12-23
PCT/EP2020/087673 WO2021130250A1 (en) 2019-12-23 2020-12-22 New mesothelin specific chimeric antigen receptors (car) for solid tumors cancer immunotherapy

Publications (1)

Publication Number Publication Date
EP4081537A1 true EP4081537A1 (de) 2022-11-02

Family

ID=74205797

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20845393.6A Pending EP4081537A1 (de) 2019-12-23 2020-12-22 Neue mesothelin-spezifische chimäre antigen-rezeptoren (car) für die krebsimmuntherapie bei soliden tumoren

Country Status (10)

Country Link
US (1) US20230068949A1 (de)
EP (1) EP4081537A1 (de)
JP (1) JP2023507525A (de)
KR (1) KR20220118532A (de)
CN (1) CN115175928A (de)
AU (1) AU2020415318A1 (de)
CA (1) CA3166356A1 (de)
IL (1) IL294118A (de)
MX (1) MX2022007833A (de)
WO (1) WO2021130250A1 (de)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113527515A (zh) * 2021-07-14 2021-10-22 南京蓝盾生物科技有限公司 一种靶向mesothelin的嵌合抗原受体及其应用
WO2023034729A1 (en) * 2021-08-30 2023-03-09 Carrygenes Bioengineering, Llc Safety switches for engineered cells carrying synthetic chromosomes
WO2024096592A1 (ko) * 2022-11-02 2024-05-10 주식회사 셀렌진 메소텔린에 대한 친화도가 상승된 키메릭 항원 수용체 및 이의 용도

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (fr) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Système d'aimant à entrefer annulaire
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1994024277A1 (en) 1993-04-13 1994-10-27 Sloan-Kettering Institute For Cancer Research Protection of human bone marrow from high dose antifolate therapy using mutated human dihydrofolate reductase dna
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
WO1997033988A1 (en) 1996-03-12 1997-09-18 Sloan-Kettering Institute For Cancer Research Double mutants of dihydrofolate reductase and methods of using same
MXPA02008265A (es) 2000-02-24 2004-09-10 Xcyte Therapies Inc Concentracion y estimulacion simultanea de calulas.
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
EP3202899B1 (de) 2003-01-28 2020-10-21 Cellectis Massgearbeitete meganuklease und verwendung davon
WO2004083379A2 (en) 2003-03-14 2004-09-30 Walters Richard E Large volume ex vivo electroporation method
WO2013063419A2 (en) * 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania A fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
WO2013176916A1 (en) 2012-05-25 2013-11-28 Roman Galetto Use of pre t alpha or functional variant thereof for expanding tcr alpha deficient t cells
WO2014004549A2 (en) * 2012-06-27 2014-01-03 Amgen Inc. Anti-mesothelin binding proteins
SG11201501471VA (en) 2012-09-04 2015-03-30 Cellectis Multi-chain chimeric antigen receptor and uses thereof
JP2016524464A (ja) 2013-05-13 2016-08-18 セレクティスCellectis 免疫療法のために高活性t細胞を操作するための方法
AU2014351797B2 (en) 2013-11-22 2021-03-11 Cellectis Method of engineering chemotherapy drug resistant T-cells for immunotherapy
RU2714902C2 (ru) * 2013-12-19 2020-02-20 Новартис Аг Химерные рецепторы антигена против мезотелина человека и их применение
PL3116902T3 (pl) 2014-03-11 2020-07-27 Cellectis Sposób wytwarzania limfocytów T kompatybilnych do przeszczepienia alogenicznego
EP3250606B1 (de) 2015-01-26 2020-11-25 Cellectis Anti-cll1-spezifische einkettige chimäere antigenrezeptoren(sccars) zur krebsimmuntherapie
WO2018073391A1 (en) 2016-10-19 2018-04-26 Cellectis Targeted gene insertion for improved immune cells therapy
CN109593721B (zh) * 2017-09-30 2022-11-01 亘喜生物科技(上海)有限公司 具有自杀基因开关的靶向人间皮素的工程化免疫细胞

Also Published As

Publication number Publication date
MX2022007833A (es) 2022-09-23
US20230068949A1 (en) 2023-03-02
WO2021130250A1 (en) 2021-07-01
IL294118A (en) 2022-08-01
CN115175928A (zh) 2022-10-11
AU2020415318A1 (en) 2022-07-14
CA3166356A1 (en) 2021-07-01
JP2023507525A (ja) 2023-02-22
KR20220118532A (ko) 2022-08-25

Similar Documents

Publication Publication Date Title
US11498971B2 (en) BCMA (CD269) specific chimeric antigen receptors for cancer immunotherapy
US11365430B2 (en) Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
AU2014273490B2 (en) Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
JP2020537528A (ja) 改善された免疫細胞療法のためのnk阻害物質遺伝子の標的指向遺伝子組み込み
AU2016212159A1 (en) Anti-HSP70 specific Chimeric Antigen Receptors (CARs) for cancer immunotherapy
US20230068949A1 (en) New mesothelin specific chimeric antigen receptors (CAR) for solid tumors cancer immunotherapy
US20230248825A1 (en) T-cells expressing immune cell engagers in allogenic settings
US11903968B2 (en) Engineered immune cells resistant to tumor microenvironment
AU2022267804A1 (en) New anti-muc1 cars and gene edited immune cells for solid tumors cancer immunotherapy
CN117580868A (zh) 用于实体瘤癌症免疫疗法的新型抗-muc1 car和基因编辑的免疫细胞

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220715

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)